Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs and Goats [11 ed.] 0702052469, 9780702052460

Treat the diseases affecting large animals! Veterinary Medicine, 11th Edition provides up-to-date information on the dis

2,890 309 47MB

English Pages 2278 [2356] Year 2016

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs and Goats [11 ed.]
 0702052469, 9780702052460

Table of contents :
Volume 1
Front Cover
Veterinary Medicine
Copyright Page
Dr. Otto M. Radostits, August 31, 1934-December 15, 2006, Senior Author, Fifth to Seventh Editions; Lead Author, Eighth to Tenth Editions
Dedictaion_Clive Gay
1920–2013
Contributors
Preface to the Eleventh Edition
Introduction
Veterinary Medicine in the Anthropocene
Contemporary Livestock Production
Veterinary Clinical Epidemiology
Veterinary Scientific Literature and How to Use It
Quality of Evidence
From Evidence to Recommendation
Food- and Fiber-Producing Animals
Industrialized Animal Agriculture
Companion-Animal Practice
Equine Practice
Contrasting Objectives
The Objectives of Food-Producing-Animal Practice
Efficiency of Livestock Production
Animal Welfare
Zoonoses and Food Safety
Principles of Food-Producing Animal Practice
Regular Farm Visits
Clinical Examination and Diagnosis
Examination of the Herd
Collection and Analysis of Animal Health Data
Public Health and Food Safety
Economics of Veterinary Practice
Veterinary Education
Optimal Utilization of the Food-Producing-Animal Practitioner
Further Reading
References
Table Of Contents
List of Tables
List of Illustrations
1 Clinical Examination and Making a Diagnosis
Introduction
Making a Diagnosis
Diagnostic Methods
Method 1: the Syndrome or Pattern Recognition
Method 2: Hypothetico-Deductive Reasoning
Method 3: the Arborization or Algorithm Method
Method 4: the Key Abnormality Method
Determination of the Abnormality of Function Present
Determination of the System or Body as a Whole or Organ Affected
Determination of the Location of the Lesion Within the System or Organ Affected
Determination of the Type of Lesion
Determination of the Specific Cause of the Lesion
Method 5: the Database Method
Clinical Examination of the Individual Animal
History Taking
History-Taking Method
Animal Data
Disease History
Present Disease
Morbidity, Case Fatality, and Population Mortality Rates
Prior Treatment
Prophylactic and Control Measures
Previous Exposure
Transit
Culling Rate
Previous Disease
Management History
Nutrition
Livestock at Pasture
Hand-Fed/Stall-Fed Animals
Reproductive Management and Performance
Breeding History
Reproductive History
Climate
General Management
Examination of the Environment
Outdoor Environment
Topography and Soil Type
Stocking Rate (Population Density)
Feed and Water Supplies
Pasture and Feed
Water
Waste Disposal
Indoor Environment
Hygiene
Ventilation
Flooring
Floor Plan
Lighting
Examination of the Animal
General Inspection (Distant Examination)
Behavior and General Appearance
Behavior
Excitation States
Voice
Eating
Defecation
Urination
Posture
Gait
Body Condition
Body Conformation
Skin
Inspection of Body Regions (Particular Distant Examination)
Head
Neck
Thorax
Respiratory Rate
Respiratory Rhythm
Prolongation of Phases
Respiratory Depth
Type of Respiration
Thorax Symmetry
Respiratory Noises or Stridors
Abdomen
External Genitalia
Mammary Glands
Limbs
Close Physical Examination
Palpation
Percussion
Ballottement
Auscultation
Percussion and Simultaneous Auscultation of the Abdomen
Succussion
Other Techniques
Sequence Used in the Close Physical Examination
Vital Signs
Temperature
Pulse
Rate
Rhythm
Amplitude
State of Hydration
Examination of Body Regions
Thorax
Cardiac Area
Lung Area
Abdomen
Auscultation
Auscultation of the Rumen of Cattle and Sheep
Intestinal Sounds of the Horse
Palpation and Percussion Through the Abdominal Wall
Percussion and Simultaneous Auscultation
Tactile Percussion of the Abdomen
Abdominal Pain
Nasogastric Intubation
Head and Neck
Eyes
Examination of the Conjunctiva
Corneal Abnormalities
Size of the Eyeball
Abnormal Eyeball Movements
Examination of the Deep Structures
Vision Tests
Nostrils
Mouth
Teeth
Tongue
Pharynx
Submaxillary Region
Neck
Rectal Examination
Feces and Defecation
Color of the Feces.
Fecal Odor.
Composition.
Frequency of Defecation.
Other Observations
Paracentesis of the Abdomen
Urinary System
Reproductive Tract
Mammary Gland
Musculoskeletal System and Feet
Nervous System
Skin Including Ears, Hooves, and Horns
Diagnostic Imaging
Further Reading
References
Interpretation of Laboratory Data
Why Collect Laboratory Data?
Properties of Diagnostic Tests
Utility
Reference Range (Interval)
Problems With Reference Ranges
Sensitivity and Specificity
Likelihood Ratio
Positive and Negative Predictive Value
Further Reading
References
Computer-Assisted Diagnosis
Reference
Prognosis and Therapeutic Decision Making
Systematic Reviews
Decision Analysis
Further Reading
References
2 Examination of the Population
Approach to Examining the Population
Examination Steps
Step 1: Defining the Abnormality
Step 2: Defining the Pattern of Occurrence and Risk Factors
Temporal Pattern
Spatial Examination
Step 3: Defining the Etiologic Group
Step 4: Defining the Specific Etiology
Techniques in Examination of the Herd or Flock
Clinical Examination
Sampling and Laboratory Testing
Numerical Assessment of Performance
Intervention Strategies and Response Trials
Role of the Integrated Animal Health and Production Management Program
Further Reading
References
3 Biosecurity and Infection Control
Definitions and Concepts
Development of a Biosecurity Plan
Initial Planning
Practices to Aid in Maintaining Biosecurity
Testing and/or Isolation of Newly Introduced Animals
Controlling Contact by Visitors to the Operation
Controlling Contact by Wildlife, Neighboring Livestock, and Pets
Separating Groups of Animals Based on Risk
Cleaning and Disinfection
Disease Monitoring and Record Keeping
Communication, Training, and Assessment
Further Reading
References
4 General Systemic States
Hypothermia, Hyperthermia, and Fever
Body Temperature
Heat Production
Heat Loss
Balance Between Heat Loss and Gain
Breed Differences
Hypothermia
Etiology
Excessive Loss of Heat
Insufficient Heat Production
Combination of Excessive Heat Loss and Insufficient Heat Production
Epidemiology
Neonatal Hypothermia
Thermoregulation in Neonatal Farm Animals
Response to Cold Stress
Cold-Induced Thermogenesis
Control of Heat Loss
Tissue Insulation.
External Insulation.
Thermoregulating Mechanisms
Heat Production
Cold Thermogenesis
Shivering Thermogenesis.
Nonshivering Thermogenesis.
Summit Metabolism.
Birth Weight and Summit Metabolism.
Factors Affecting Cold Thermogenesis
Malnutrition of the Dam During Late Gestation.
Postnatal Changes in Cold Thermogenesis
Risk Factors for Neonatal Hypothermia
Calves
Lambs
Piglets
Foals
Postshearing Hypothermia in Sheep
Cold Environments and Animal Production
Pathogenesis
Clinical Findings
Neonatal Hypothermia
Shorn Sheep Hypothermia
Hypothermia Secondary to Other Diseases
Clinical Pathology
Necropsy Findings
Treatment
Hypothermic Newborn Lambs
Hypothermic Newborn Calves
Hypothermic Newborn Foals
Hypothermic Newborn Piglets
Control
Lambs and Calves
Piglets
Sick Foals
Further Reading
Reference
Cold Injury (Frostbite and Chilblains)
Etiology and Epidemiology
Pathogenesis
Clinical Findings
Necropsy Findings
Treatment
Control
Hyperthermia (Heat Stroke or Heat Exhaustion)
Etiology
High Environmental Temperature
Other Causes of Hyperthermia
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Fever (Pyrexia)
Etiology
Septic Fevers
Aseptic Fevers
Pathogenesis
Endogenous Pyrogens
Effect of Pyrogens on the Hypothalamus
Febrile Response
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Antimicrobial Agents
Antipyretics
Further Reading
Acute Phase Response
Further Reading
References
Sepsis, Septicemia, and Viremia
Etiology: All Species
Neonatal Septicemias
Calves
Piglets
Foals
Lambs
Cattle
Sheep (Young Lambs)
Pigs
Horses, Donkeys, and Mules
Secondary Septicemias
Epidemiology
Pathogenesis
Disseminated Intravascular Coagulation
Clinical Findings
Neonatal Septicemia
Clinical Sepsis Score
Clinical Pathology
Blood Culture
Hemogram
Immunoglobulin Status
Biomarkers
Serology
Necropsy Findings
Treatment
Further Reading
References
Toxemia, Endotoxemia, and Septic Shock
Etiology
Antigenic Toxins
Exotoxins
Enterotoxins
Endotoxins
Metabolic Toxins
Pathogenesis
Biochemical Mediators
Cardiopulmonary Function
Activation of the Renin–Angiotensin–Aldosterone System and Dysfunction of the Hypothalamic–Pituitary–Adrenal Axis
Leukocytes and Platelets
Hemostatic System
Thermoregulation
Gastrointestinal Function
Carbohydrate Metabolism
Protein Metabolism
Mineral Metabolism
Reproduction and Lactogenesis
Combined Effects on Body Systems
Endotoxin Tolerance
Hypersensitivity
Other Infectious Toxins
Clinical Findings
Acute Toxemia
Endotoxemia
Chronic Toxemia
Localized Infection
Clinical Pathology
Hematology
Serum Biochemistry
Endotoxin
Necropsy Findings
Treatment
Removal of Foci of Infection
Antimicrobial Agents
Aggressive Fluid Therapy
Hypertonic Solutions
Glucose and Insulin Administration
Inotropic Agents, Vasopressors, and Local Anesthetics
Nonsteroidal Antiinflammatory Drugs
Glucocorticoids
Polymyxin B
Hyperimmune Serum and Plasma Transfusion
Pentoxifylline and Ethyl Pyruvate
Anticoagulants
Control of Endotoxemia
Further Reading
References
Toxemia in the Recently Calved Cow
Puerperal Metritis in Cattle
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Hematology
Vaginal/Uterine Fluid
Other Samples and Tests
Necropsy Findings
Fat Cow Syndrome
Acute Diffuse Peritonitis
Peracute and Acute Mastitis
Treatment
Conservative Therapy
Antimicrobial Agents
Intrauterine Medication
Ancillary Treatment and Control
Identification of Affected Cows
Further Reading
References
Hypovolemic, Hemorrhagic, Maldistributive, and Obstructive Shock
Etiology
Hypovolemic Shock
Hemorrhagic Shock
Cattle, Sheep, and Goats
Horses
Pigs
Maldistributive Shock
Obstructive Shock
Pathogenesis
Hypovolemic Shock
Hemorrhagic Shock
Maldistributive Shock
Obstructive Shock
Clinical Findings
Clinical Pathology
Monitoring in Shock
Necropsy Findings
Treatment
Identification of Cause
Hypovolemic and Maldistributive Shock
Isotonic Crystalloid Solutions
Hypertonic Saline Solution
Colloids
Hemorrhagic Shock
Obstructive Shock
Ancillary Treatment
Corticosteroids
Cyclooxygenase Inhibitors
Antibiotic Therapy
Vasoconstrictors and Vasodilators
Immunotherapy
Further Reading
References
Localized Infections
Etiology
Bacterial Causes of Localized Infection
Portal of Entry
Pathogenesis
Clinical Findings
Clinical Pathology
Hemogram
Sample of Lesion for Culture and Staining
Necropsy Findings
Treatment
Drainage of Abscesses
Antimicrobial Agents
References
Pain
The Problem of Pain
Advances in Attitude Toward Pain
Etiology
Cutaneous or Superficial Pain
Visceral Pain
Musculoskeletal (Somatic) Pain
Pathogenesis
Central Hypersensitivity and Preemptive Analgesia
Clinical Findings
Physiologic Responses
Behavioral Responses
Elicitation of Pain by the Veterinarian
Periodicity and Duration of Pain
Treatment
Analgesia
Analgesic Agents
Local Anesthetic Agents
Nonsteroidal Antiinflammatory Drugs
Flunixin Meglumine
Ketoprofen
Phenylbutazone
Meloxicam
Salicylates
Carprofen
Diclofenac
α2-Agonists
Xylazine
Narcotic Analgesics
N-methyl-d-aspartate receptor antagonists
Vanilloids
γ-Aminobutyric Acid Analogs
Balanced (Multimodal) Analgesia
Administration Routes
Supportive Therapy
Further Reading
References
Stress
Causes of Stress
Pathogenesis
Stress and Road Transportation
Other Possible Sources of Stress
Clinical Pathology
Stress Syndromes
Stress-Related Psychosomatic Disease
Stress and Susceptibility to Infection
Stress and Animal Welfare
Stress and Metabolic Disease
Stress and Its Effect on Economic Performance
Management of Stress
Further Reading
References
Disturbances of Appetite, Food Intake, and Nutritional Status
Thirst
Polyphagia
Anophagia or Aphagia
Further Reading
Reference
Pica or Allotriophagia
Cannibalism
Infantophagia
Significance of Pica
Starvation
Inanition (Malnutrition)
Further Reading
References
Weight Loss or Failure to Gain Weight (Ill-Thrift)
Nutritional Causes
Excessive Loss of Protein and Carbohydrates
Faulty Digestion, Absorption, or Metabolism
Shortfalls in Performance
Further Reading
References
Unthriftiness in Weaner Sheep (Weaner Ill-Thrift)
Etiology
Epidemiology
Clinical and Necropsy Findings
Further Reading
References
Porcine Failure to Thrive
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Treatment
Control
References
Physical Exercise and Associated Disorders
Further Reading
Exercise-Associated Diseases
Exhaustion
Pathogenesis
Clinical Signs
Treatment
Prevention
References
Poor Racing Performance and Exercise Intolerance in Horses
Approach to the Horse With Exercise Intolerance
History
Clinical Examination
Laboratory Testing
Exercise Stress Testing
Causes of Exercise Intolerance or Poor Performance
Musculoskeletal System
Cardiovascular System
Respiratory System
Upper Airways (See Obstructive Diseases of the Equine Larynx)
Lower Airways
Hematologic and Biochemical Abnormalities
Anemia
Hypoproteinemia
Electrolyte Abnormalities
Nervous System Disease
Miscellaneous
Treatment
Further Reading
References
Sudden or Unexpected Death
Sudden or Unexpected Death in Single Animals
Spontaneous Internal Hemorrhage
Rupture of Internal Carotid Artery Aneurysm
Peracute Endogenous Toxemia
Transportation Stress
Trauma
Gastrointestinal Conditions
Iatrogenic Deaths
Sudden Death in Horses
Sudden or Unexpected Death in a Group of Animals
Lightning Strike or Electrocution
Nutritional Deficiency and Poisoning
Access to Potent Poisons
Diseases Associated With Infectious Agents
Neonatal and Young Animals
Anaphylaxis
Procedure for Investigation of Sudden Death
Further Reading
References
Cyanobacteria (Blue-Green Algae) Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Pathogenesis
Microcystins
Anatoxins
Anatoxin-a
Homoanatoxin-a
Anatoxin-a(s)
Various Freshwater Toxins
Clinical Findings
Microcystins
Anatoxins
Clinical Pathology
Microcystins
Anatoxins
Necropsy Findings
Diagnosis
Treatment
Control
Prevent the Ingestion of Toxins
Prevent the Addition of Nutrients to Water
Further Reading
References
Plants Causing Sudden Death Without Cardiomyopathy
Diseases Associated With Physical Agents
Lightning Strike and Electrocution
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnosis
Treatment
Control
Further Reading
References
Stray Voltage
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Behavioral Changes
Effects on Production and Disease
Further Reading
References
Environmental Pollutants and Noise
Pollution From Outside the Farm
Pollution From Farms
Noise
References
Wind Farms and Electric and Magnetic Fields
Further Reading
Radiation Injury
Etiology
Epidemiology
Incidence and Case Fatality
Risk Factors
Animal
Nature of Radiation
Zoonotic Implications
Pathogenesis
Clinical Findings
Acute Syndrome
Subacute Syndrome
Chronic Exposure
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Control
References
Volcanic Eruptions
Blast and Gas Damage
Ash Hazards
Toxic Chemicals
Physical Properties
Long-Term Effects
References
Bushfire (Grass fire) Injury (Thermal Burns)
Etiology
Epidemiology
Forest Fires
Grass Fires
Barn Fires
Clinical Findings
Burn Injury
Treatment
Decision Criteria
Skin Burns
Smoke Inhalation
Further Reading
Diagnosis of Inherited Disease
Diagnosis
Control of Inherited Disease
Online Mendelian Inheritance in Animals
References
5 Disturbances of Free Water, Electrolytes, Acid-Base Balance, and Oncotic Pressure
Dehydration
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Water Intoxication
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Postmortem Findings
Treatment
Control
Further Reading
References
Electrolyte Imbalances
Hyponatremia
Hypernatremia
Hypochloremia
Hypokalemia
Hyperkalemia
Hypocalcemia
Hypophospatemia
Hypomagnesemia
Further Reading
References
Acid-Base Imbalance
Acidemia
Etiology
Pathogenesis
Clinical Findings
Alkalemia
Etiology and Pathogenesis
Clinical Findings
Oncotic Pressure and Edema
Etiology
Decreased Plasma Oncotic Pressure
Increased Hydrostatic Pressure
Increased Capillary Permeability
Obstruction to Lymphatic Flow
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Naturally Occurring Combined Abnormalities of Free Water, Electrolyte, Acid-Base Balance, and Oncotic Pressure
Nature of the Disease and History
Clinical Findings
Clinical Pathology
Packed Cell Volume and Total Serum Protein or Plasma Protein
Total CO2
Blood Gas and pH Analysis
Blood or Plasma l-Lactate Concentration
Serum Electrolytes
Urea and Creatinine
Blood or Plasma Glucose
Anion Gap
Strong Ion Gap
Osmolal Gap
Arterial, Jugular, or Central Venous Blood Pressure
Total Body Water
Further Reading
References
Principles of Fluid and Electrolyte Therapy
Calculation of Electrolyte Requirements
Types of Intravenous Fluid
Crystalloid Solutions
Isotonic, Hypertonic, and Hypotonic Crystalloid Solutions
Hypotonic Crystalloid Solutions
Isotonic Crystalloid Solutions
Hypertonic Crystalloid Solutions
Colloid Solutions
Practical Administration of Electrolyte Solutions
Quantity of Fluids Required and Routes of Administration
Parenteral Fluid Therapy
Rate of Administration
Intravenous Catheters and Complications
Auricular Vein of Cattle and Calves
Cecal Catheters in Horses
Thrombophlebitis
Oral Fluid Therapy
Oral Fluid Therapy in Calves and Adult Cattle
Oral Fluid Therapy in Horses
Fluid and Electrolyte Therapy in Newborn Piglets and Lambs
Parenteral Nutrition
Further Reading
References
6 Practical Antimicrobial Therapeutics
Principles of Antimicrobial Therapy
Identification of the Infection by Clinical Examination
Taking Samples for Diagnosis
Antimicrobial Susceptibility Tests
Rationale
Susceptibility Test Methods
Tube Susceptibility Tests
Disk Susceptibility Tests
Microtiter Techniques
Other Considerations
Flaws and Limitations of Culture Susceptibility Testing
Further Reading
Antibiotic Resistance
Further Reading
Ways to Minimize or Avoid the Development of Antimicrobial Resistance
Avoiding Antimicrobial Resistance (Three Ds Approach)
Antibiotic Metaphylaxis to Control Respiratory Disease
Further Reading
Practical Usage of Antimicrobial Drugs
Antibiotic Dosage: the Recommended Dose
Routes of Administration
Intravenous Injection
Intramuscular Injection
Intraperitoneal Injection
Subcutaneous Injection
Oral Administration
Contamination of Feedstuffs
Water Medication of Pigs
Water Medication in Cattle
Dietary Medication
Ionophores
Other Routes
Further Reading
Drug Distribution
Absorption
Distribution
Pharmacokinetic Principles for Antimicrobial Usage
Duration of Treatment
Drug Combinations
Further Reading
Additional Factors Determining Selection of Agents
Cost
Ease of Administration
Toxicity
Bactericidal or Bacteriostatic Antimicrobials
Postantibiotic Effect
Concentration-Dependent Killing
Time-Dependent Killing
Drug Deterioration
Unfavorable Response to Therapy
Drug Withdrawal Requirements and Residue Avoidance
Label Dose and Extralabel Use
Extralabel Use
Withdrawal Periods
Residue Testing
Testing for Compliance
Causes of Residue Violations in Milk
Causes of Residue Violations in Beef Cattle
Causes of Residue Violations in Swine
Type of Therapy
Approved Drugs
Classification of Antimicrobial Agents: Mechanisms of Action and Major Side Effects
Aminoglycosides and Aminocyclitols
Mechanism of Action
Toxicity
β-Lactam Antibiotics: Penicillins, Cephalosporins, and β-Lactamase Inhibitors
Mechanism of Action
Toxicity and Clinical Considerations
Toxicity
Chloramphenicol
Mechanism of Action
Toxicity
Chloramphenicol Analogs
Toxicity
Fluoroquinolones
Mechanism of Action
Toxicity
Lincosamides
Mechanism of Action
Toxicity
Macrolides
Mechanism of Action
Toxicity
Sulfonamides
Mechanism of Action
Toxicity and Clinical Considerations
Precautions and Contraindications
Tetracyclines
Mechanism of Action
Toxicity
Miscellaneous Antibiotics
Bacitracin
Carbadox
Dapsone
Metronidazole
Nitrofurans (e.g., Nitrofurantoin, Nitrofurazone, Furazolidone)
Novobiocin
Polymyxins
Rifampin
Vancomycin
Virginiamycin
Further Reading
7 Diseases of the Alimentary Tract
Principles of Alimentary Tract Dysfunction
Motor Function
Normal Gastrointestinal Motility
Hypermotility and Hypomotility
Distension
Abdominal Pain
Dehydration and Shock
Secretory Function
Digestive Function
Absorptive Function
Manifestations of Alimentary Tract Dysfunction
Abnormalities of Prehension, Mastication, and Swallowing
Causes of Dysphagia and Inability to Swallow
Drooling of Saliva and Excessive Salivation
Local Causes of Drooling
Systemic Causes of Excessive Salivation
Vomiting and Regurgitation
Vomiting
Projectile Vomiting
True Vomiting
Regurgitation
Diarrhea, Constipation, and Scant Feces
Diarrhea
Malabsorption Syndromes
Constipation
Scant Feces
Ileus (Adynamic and Dynamic Ileus)
Alimentary Tract Hemorrhage
Abdominal Pain
Horses
Cattle
Common Causes of Alimentary Tract Pain
Horses
Cattle
Tenesmus
Cattle
Horses
Pigs
Shock and Dehydration
Abdominal Distension
Abnormal Nutrition
Special Examination
Nasogastric Intubation
Rumen of Cattle
Decompression of Distended Rumen
Decompression of the Horse’s Stomach
Medical Imaging
Radiography
Abdominal Ultrasonography
Horse
Cattle
Endoscopy
Laparoscopy
Exploratory Laparotomy (Celiotomy)
Tests of Digestion and Absorption
Glucose Absorption Test
Starch Digestion Test
Lactose Digestion Test
Xylose Absorption Test
Sucrose Absorption Test
Radioactive Isotopes
Abdominocentesis for Peritoneal Fluid
Equine and Bovine Peritoneal Fluid
Specific Properties of Peritoneal Fluid (Normal and Abnormal)
Color
Cellular and Other Properties
Cells
Abdominocentesis in Horses
Risks
Abdominocentesis in Cattle
Intestinal and Liver Biopsy
Principles of Treatment in Alimentary Tract Disease
Relief of Abdominal Pain
Relief of Distension
Replacement of Fluids and Electrolytes
Correction of Abnormal Motility
Increased Motility
Decreased Motility
Metoclopramide
Cisapride
Xylazine and Naloxone
Bethanechol and Neostigmine
Relief of Tenesmus
Reconstitution of Rumen Flora and Correction of Acidity OR Alkalinity
Further Reading
References
Diseases of the Buccal Cavity and Associated Organs
Diseases of the Muzzle
Stomatitis
Etiology
Physical Agents
Chemical Agents
Infectious Agents
Cattle
Sheep
Horses
Pigs
Bullous Stomatitis
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Diseases of the Teeth
Etiology
Congenital Defects
Dental Fluorosis
Enamel Erosion
Premature Wear and Loss of Teeth in Sheep (Periodontal Disease)
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment and Control
References
Diseases of the Parotid Salivary Glands
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Diseases of the Pharynx and Esophagus
Pharyngitis
Etiology
Physical Causes
Infectious Causes
Cattle
Horses
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Pharyngeal Obstruction
Etiology
Foreign Bodies
Tissue Swellings
Cattle
Horses
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Pharyngeal Paralysis
Etiology
Peripheral Nerve Injury
Secondary to Specific Diseases
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Reference
Esophagitis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Esophageal Rupture
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Esophageal Obstruction
Etiology
Intraluminal Obstructions
Extraluminal Obstructions
Esophageal Paralysis
Megaesophagus
Esophageal Strictures
Other Causes of Obstruction
Pathogenesis
Clinical Findings
Acute Obstruction or Choke
Cattle
Horse
Chronic Obstruction
Complications Following Esophageal Obstruction
Clinical Pathology
Treatment
Conservative Approach
Sedation
Pass a Stomach Tube and Allow Object to Move Into Stomach
Removal by Endoscope
Manual Removal Through Oral Cavity in Cattle
General Anesthesia in the Horse
Esophageal Lavage in the Horse
Surgical Removal of Foreign Bodies
Repeated Siphonage in Chronic Cases
Cervical Esophagostomy Alimentation
Antimicrobial Administration
References
Diseases of the Nonruminant Stomach and Intestines
Gastritis
Etiology
Cattle and Sheep
Physical Agents
Chemical Agents
Infectious Agents
Metazoan Agents
Pigs
Physical Agents
Chemical Agents
Infectious Agents
Metazoan Agents
Horses
Pathogenesis
Clinical Findings
Acute Gastritis
Chronic Gastritis
Clinical Pathology
Necropsy Findings
Treatment
Enteritis (Including Malabsorption, Enteropathy, and Diarrhea)
Etiology and Epidemiology
Pathogenesis
Normal Intestinal Absorption
Mechanisms of Diarrhea
Osmotic Diarrhea
Exudative Diarrhea
Secretory Diarrhea
Abnormal Intestinal Motility
Location of Lesion
Dehydration, Electrolyte, and Acid-Base Imbalance
Chronic Enteritis
Replacement of Villous Epithelial Cells
Role of Neutrophils in Intestinal Mucosal Injury
Intestinal Motility in Enteritis
Concurrent Gastritis
Effects of Enteritis on Pharmacodynamics of Drugs
Clinical Findings
Systemic Effects
Intestinal Sounds in Enteritis
Chronic Enteritis
Clinical Pathology
Fecal Examination
Intestinal Tissue Samples
Hematology and Serum Biochemistry
Digestion/Absorption Tests
Necropsy Findings
Treatment
Removal of Causative Agent
Antimicrobials
Mass Medication of Feed and Water Supplies
Alteration of the Diet
Fluids and Electrolytes
Intestinal Protectants and Adsorbents
Antidiarrheal Drugs
Antimotility Drugs
Antisecretory Drugs
Control
Intestinal Hypermotility
Dietary Diarrhea
Etiology
Milk Replacers
Overfeeding of Milk
Change of Diet
Pathogenesis
Digestion of Milk
Milk Replacers and Diarrhea
Clinical Findings
Nursing Beef Calves
Hand-Fed Dairy Calves
Milk-Replacer Diarrhea
Clinical Pathology
Necropsy Findings
Treatment
Alter Diet of Hand-Fed Calves
Monitor Beef Calves With Dietary Diarrhea
Reference
Abdominal FAT Necrosis (Lipomatosis)
References
Diseases of the Peritoneum
Peritonitis
Etiology
Cattle
Horses
Pigs
Sheep
Goats
All Species
Pathogenesis
Toxemia and Septicemia
Shock and Hemorrhage
Abdominal Pain
Paralytic Ileus
Accumulation of Fluid Exudate
Adhesions
Clinical Findings
Acute and Subacute Peritonitis
Inappetence and Anorexia
Toxemia and Fever
Feces
Alimentary Tract Stasis
Abdominal Pain Evidenced by Posture and Movement
Abdominal Pain as Evidenced by Deep Palpation
Rectal Examination
Peracute Diffuse Peritonitis
Chronic Peritonitis
Cattle
Horses
Diagnostic Medical Imaging
Clinical Pathology
Hematology
Abdominocentesis and Peritoneal Fluid
Septic Peritonitis in the Horse
Peritonitis in Cattle
Necropsy Findings
Diagnosis
Prognosis
Case–Fatality Rate in Horses
Treatment
Antimicrobials
Fluid and Electrolytes
Nonsteroidal Antiinflammatory Drugs
Lavage
Prevention of Adhesions
References
Abdominal Diseases of the Horse Including Colic and Diarrhea
General Principles
Etiology
Epidemiology
Occurrence
Risk Factors
Horse Characteristics
Age
Sex
Breed
Diet and Feeding Practices
Management
Watering
Housing
Exercise
Season and Weather
Medical History
Parasite Control
Importance
Pathogenesis
Pain
Gastrointestinal Dysfunction
Ischemia of the Intestinal Wall
Endotoxemia
Shock
Coagulation and Fibrinolysis
Overview of the Pathogenesis of Common Colics
Simple Obstructive
Obstructive and Strangulating
Infarctive
Inflammatory
Clinical Findings
Visual Examination
Behavior
Posture
Abdomen Size
Vomiting
Defecation and Feces
Physical Examination
Heart and Respiratory Rates
Mucous Membranes and Extremities
Auscultation and Percussion
Rectal Examination
Normal Anatomy
Abnormal Findings
Nasogastric Intubation
Ancillary Diagnostic Techniques
Ultrasonography
Radiology
Arterial Blood Pressure
Course of the Disease
Clinical Pathology
Hematology and Serum Biochemistry
Acid-Base Status
Abdominocentesis
Protocol for Evaluating a Colic Patient
Behavior
Clinical and Clinicopathologic Observations
When to Refer the Patient
Surgery
Prognosis
Necropsy Findings
Treatment
Medical Treatment
Analgesia
Nonsteroidal Antiinflammatory Drugs
α-2 Agonists
Opiates
Other Agents
Prophylaxis and Treatment of Endotoxemia
Fluid and Electrolyte Therapy
Intestinal Lubricants and Fecal Softeners
Other Treatments
Trocarization
Management of Field Colic
Surgery
Prevention
Further Reading
References
Colic in the Pregnant and Postparturient Mare
Further Reading
References
Colic in Foals
Etiology
Epidemiology
Pathophysiology
Clinical Findings
Ancillary Diagnostic Tests
Diagnostic Imaging
Endoscopy
Clinical Pathology
Necropsy Findings
Treatment
Surgical Treatment
Prevention
Further Reading
References
Gastric Dilation in the Horse
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Gastric Impaction IN Horses
Further Reading
References
Gastric (Gastroduodenal) Ulcer in Foals
Etiology
Epidemiology
Occurrence
Risk Factors
Age and Sex
Stress and Disease
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
References
Gastric Ulcer in Adult Horses
Etiology
Epidemiology
Occurrence
Lesions of the Squamous Versus Glandular Mucosa
Risk Factors
Animal Risk Factors
Management and Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Acid Suppression
Omeprazole
Cimetidine
Ranitidine and Famotidine
Gastric Antacids
Protectants and Other Treatments
Management Changes
Overview of Treatment
Control
Further Reading
References
Intestinal Obstruction IN Horses
Small-Intestinal Obstruction in Horses
Etiology
Obstruction With Infarction
Obstruction Without Infarction
Functional Obstruction
Epidemiology
Intestinal Herniation Through the Epiploic Foramen
Pedunculated Lipomas
Inguinal Hernias
Intussusception
Foreign Body
Impaction
Mesenteric Rents
Pathogenesis
Clinical Findings
Acute Disease: Infarctive Lesions
Subacute Cases: Noninfarctive Lesions
Intussusception of the Small Intestine
Volvulus of the Small Intestine
Strangulated Inguinal Hernia
Strangulated Diaphragmatic Hernia
Epiploic Foramen Entrapment
Functional Obstruction
Foreign Body
Ileocecal Valve Impaction
Idiopathic Muscular Hypertrophy (Terminal Ileal Hypertrophy)
Caudal Abdominal Obstructions
Clinical Pathology
Necropsy Findings
Treatment
References
Duodenitis-Proximal Jejunitis (Anterior Enteritis, Proximal Enteritis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
References
Diseases of the Cecum
Etiology
Epidemiology
Cecal Impaction
Cecal Perforation or Rupture
Cecocecal or Cecocolic Intussusceptions
Cecal Torsion
Pathogenesis
Clinical Findings
Cecal Distension and Impaction
Perforation and Rupture
Intussusception
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Displacement and/or Volvulus of the Large (Ascending) Colon
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Left Dorsal Displacement (Renosplenic Entrapment)
Right Dorsal Displacement
Volvulus
Clinical Pathology
Necropsy Findings
Treatment
Left Displacement
Right Dorsal Displacement and Colon Volvulus
References
Impaction of the Large (Ascending) Colon of Horses
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Enteroliths and Fecaliths
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Sand Colic
Further Reading
References
Right Dorsal Colitis
Further Reading
References
Small Colon Obstruction
Epidemiology
Pathogenesis
Clinical Findings
Nonstrangulating Lesions
Strangulating Lesions
Clinical Pathology
Necropsy Findings
Treatment
Small-Colon Impaction
Further Reading
References
Spasmodic Colic
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Treatment
Intestinal Tympany in Horses
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Verminous Mesenteric Arteritis (Verminous Aneurysm and Thromboembolic Colic)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Retroperitoneal Abscess (Internal Abdominal Abscess, Chronic Peritonitis, and Omental Bursitis)
Treatment
Reference
Rectal Tears
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Prognosis
Treatment
Immediate Care
Grade I and II Tears
Grade III Tears
Grade IV Tears
Prevention
References
Acute Diarrhea of Suckling Foals
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Diagnostic Confirmation
Lesions
Treatment
Control
Further Reading
References
Acute Diarrhea of Adult (Nonsuckling) Horses
Etiology
Epidemiology
Occurrence
Risk Factors
Stress
Celiotomy
Antibiotic Administration
Pathogenesis
Clinical Signs
Clinical Pathology
Diagnostic Confirmation
Necropsy
Treatment
Restoration of Hydration
Electrolyte and Acid-Base Status
Antimicrobial Therapy
Prophylaxis and Treatment of Endotoxemia/Toxemia and Systemic Inflammatory Response
Binding of Toxins
Disseminated Intravascular Coagulation
Control
Further Reading
References
Chronic Undifferentiated Diarrhea of Horses
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Specific Treatments
Symptomatic and Supportive Treatments
Control
References
Idiopathic Chronic Inflammatory Bowel Diseases of Horses
References
Granulomatous Enteritis of Horses
Further Reading
References
Lymphocytic-Plasmacytic Enterocolitis
Further Reading
Idiopathic Focal Eosinophilic Enteritis
References
Equine Grass Sickness (Equine Dysautonomia, Grass Disease, and Mal Secco)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pasture and Soil Risk Factors
Farm or Premise Risk Factors
Transmission
Pathogenesis
Clinical Findings
Acute Cases
Subacute Cases
Chronic Cases
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Intestinal Hyperammonemia
Further Reading
References
Abdominal Diseases of the Pig Including Diarrhea
Acute Gastric Dilatation IN Pigs
Acute Gastric Volvulus IN Sows
Gastric Ulcers and Hyperkeratosis of Swine
Etiology
Epidemiology
Occurrence
Risk Factors
Dietary Risk Factors
Finely Ground Feed
Environmental and Management Risk Factors
Pathogen Risk Factors
Gastric Bacteria
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Noninfectious Intestinal Disease of Swine
Intestinal Reflux
Diagnosis
Intestinal Obstruction IN Pigs
Etiology
Clinical Findings
Impaction of the Large Intestine of Pigs
Etiology
Clinical Findings
Intestinal Tympany IN Pigs
Etiology
Osseus Metaplasia
Intestinal Hemorrhage Syndrome
Etiology
Epidemiology
Clinical Signs
Pathology
Treatment
Control
Reference
Diverticulitis and Ileitis of Pigs
Rectal Prolapse
Rectal Prolapse IN Pigs
Etiology
Epidemiology
Clinical Findings
Pathology
Treatment
Control
Rectal Stricture
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Treatment
Bacterial and Viral Diseases of the Alimentary Tract
Salmonellosis in Swine (Paratyphoid)
Etiology
Epidemiology
Prevalence and Occurrence of Infection
Belgium
Canada
Czechoslovakia
Denmark
Italy
Japan
The Netherlands
Spain
Sweden
Switzerland
Thailand
United Kingdom
United States
Morbidity and Case–Fatality Rate
Methods of Transmission and Sources of Infection
Shedding and the Carrier State
Risk Factors Predisposing to Clinical Disease
Animal Risk Factors
Genetic Resistance to Salmonellosis in Domestic Animals
Salmonella Choleraesuis
Subclinical Infections
Immune Mechanisms
Environmental and Management Risk Factors
Farming Practice in General
Housed Animals
Contaminated Feedstuffs
Introduction of the Infection to a Farm
Pathogen Risk Factors
Antimicrobial Resistance of Salmonella
Zoonotic Implications From Pigs
Salmonella Serovar Typhimurium DT104
Economic Importance
Pathogenesis
Infection
Septicemia, Bacteremia, and the Carrier State
Enteritis
Clinical Findings
Porcine Salmonellosis
Septicemia
Acute Enteritis
Chronic Enteritis
Clinical Pathology
Bacterial Culture and Detection
Fecal Culture
Multiple Fecal Cultures
DNA Probes
Serology
Serum Enzyme-Linked Immunosorbent Assay
Indirect Tests
Herd Diagnosis
Detection of Clinically Normal Carrier Animals
Determination of Prevalence of Infection in Population of Animals
Necropsy Findings
Septicemia
Acute Enteritis
Chronic Enteritis
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Primary Treatment: Antimicrobial Therapy
Control
Preventing Infection From Entering the Herd (Biosecurity)
Increased Hygiene to Prevent Intraherd Spread
Reducing Exposure to Pathogens
Avoiding Increasing Susceptibility to Pathogens
Monitoring Effectiveness of the Infectious Disease Control Program
Feed Interventions to Aid the Pigs’ Defenses
Physical Form of the Feed
Organic Acids
Feed Withdrawal
Other Options
Immunization
Salmonella Vaccinology
Monitoring
Nationwide Surveillance and Control Programs
Feedstuffs
Breeder and Multiplier Herds
Weaner Producers
Slaughter Pigs
Slurry
At Abattoir
References
Intestinal Clostridiosis in the Pig
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Differential Diagnosis
Laboratory Diagnosis
Bacteriology
Histology
Treatment
Control
References
Escherichia coli Infections in Weaned Pigs
Enterotoxigenic Escherichia coli
Verotoxin-Producing Escherichia coli
Attaching and Effacing Escherichia coli
O157 in Pigs
Edema Disease (Gut Edema, Escherichia Coli Enterotoxemia)
Etiology
Inheritance of Susceptibility to ED
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Postweaning Diarrhea of Pigs (Coliform Gastroenteritis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Resistance and E. Coli
Control
Further Reading
References
Campylobacteriosis in Pigs
Etiology
Epidemiology
Prevalence of Infection
Risk Factors
Transmission
Pathogen Risk Factors
Antimicrobial Resistance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnosis
Treatment
Control
Further Reading
References
Porcine Proliferative Enteropathy
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Morbidity and Case Fatality
Risk Factors
Methods of Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Diagnosis
Treatment and Control
Antimicrobials
Vaccines
References
Brachyspiral Colitis (Swine Dysentery, Porcine Spirochetal Colitis) and Nonspecific Colitis
Swine Dysentery
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogen Factors
Environmental and Management Risk Factors
Methods of Transmission
Pathogenesis
Clinical Findings
Immunity
Clinical Pathology
Detection and Culture of Organism
Serologic Tests
Necropsy Findings
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Choice of Antimicrobials
Failure to Respond to Therapy
Cleaning and Disinfection
Control
Control of Infection/Limitation of Reinfection
Mass Medication and Sanitation Program Without Depopulation
Dietary Modification
Depopulation and Repopulation
Biosecurity
Vaccines
References
Brachyspira Hampsonii
References
Nonspecific Colitis in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Treatment
Control
References
Porcine Intestinal Spirochetosis (Spirochetal Colitis, Porcine Colitis, and Porcine Colonic Spirochetosis) and Nonspecific Colitis
Introduction
Etiology
Occurrence
Risk Factors
Transmission
Managemental and Environmental Factors
Feed
Pathogenesis
Clinical Findings
Pathology
Immunology
Laboratory Diagnosis
Treatment
Control
Vaccination
References
Yersiniosis in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Treatment
Control
Reference
Viral Diarrhea in Neonatal Pigs
Overview of Viral Diarrhea
References
Porcine Rotaviruses
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Immunity
Treatment
Control
References
Porcine Hemagglutinating Encephalomyelitis Virus
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Differential Diagnosis
Treatment and Control
References
Porcine Adenoviruses
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Porcine Calciviruses
References
Porcine Sapoviruses
References
Porcine Norovirus
References
Porcine Astroviruses
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Diagnosis
Treatment and Control
References
Porcine Torovirus
References
Porcine Orbiviruses
Porcine Picobirnaviruses
Reference
Porcine Kobuviruses
References
Porcine New Virus
References
Porcine Bocavirus
References
A New Neonatal Diarrhea Syndrome
References
Transmissible Gastroenteritis in Pigs
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Morbidity and Case Fatality
Risk Factors
Animal Risk Factors
Level of Herd Immunity.
Herd Size.
Environmental and Management Risk Factors
Pathogen Risk Factors
Porcine Respiratory Coronaviruses
Methods of Transmission
Immune Mechanisms
Economic Losses
Pathogenesis
Clinical Findings
Piglets
Older Pigs
Clinical Pathology
Serum Biochemistry
Detection of the Virus
DNA Probe
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Control During and After an Outbreak
Isolation of Sows Due to Farrow
Discontinuation of Breeding Stock Sales and Purchases
Partial Depopulation and Culling
Planned Exposure to Virulent Virus
Biosecurity and Acquisition of Replacement Breeding Stock
All-in, All-out Management System
Complete Depopulation and Repopulation or Establishment of New Herd
TGEv Vaccines and Vaccination
Vaccination of Pregnant Sows
TGE Vaccines
Vaccine Schedule
Subunit TGEv Vaccine
Immunity to PRCV
References
Porcine Epidemic Diarrhea
Etiology
Epidemiology
Occurrence
Pathogenesis
Immunity
Clinical Signs
Pathology
Diagnosis
Treatment
Control
Further Reading
References
Swine Vesicular Disease
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Methods of Transmission
Risk Factors
Pathogen Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Vesicular Exanthema of Swine
Etiology
Epidemiology
Occurrence
Methods of Transmission
Risk Factors
Pathogen Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Salmonellosis in Ruminants and Horses
Etiology
Epidemiology
Prevalence of Infection
Cattle
Sheep
Horses
Occurrence
Cattle
Sheep
Horses
Morbidity and Case Fatality
Methods of Transmission
Carrier State
Risk Factors Predisposing to Clinical Disease
Animal Risk Factors
Genetic Resistance to Salmonellosis in Domestic Animals
Dairy Cattle
Beef Cattle Herds and Feedlots
Sheep
Horses
Immune Mechanisms
Environmental and Management Risk Factors
Cattle
Sheep
Horses
Contaminated Feedstuffs
Pathogen Risk Factors
Antimicrobial Resistance of Salmonella
Zoonotic Implications
Salmonella Typhimurium DT104
Economic Importance
Pathogenesis
Infection
Septicemia, Bacteremia, and the Carrier State
Enteritis
Abomasitis
Abortion
Terminal Dry Gangrene, Osteitis, and Polyarthritis
Clinical Findings
Septicemia
Acute Enteritis
Chronic Enteritis
Bovine Salmonellosis
Ovine and Caprine Salmonellosis
Equine Salmonellosis
Clinical Pathology
Bacterial Culture
Preenrichment Media
Enrichment Media
Selective Plating Media
DNA Recognition and Immunologic Methods
Serology
Serum Enzyme-Linked Immunosorbent Assay
Laboratory Diagnosis in a Suspected Sick Animal
Herd Diagnosis
Detection of Clinically Normal Carrier Animals
Determination of Prevalence of Infection in Population of Animals
Necropsy Findings
Septicemia
Acute Enteritis
Chronic Enteritis
Samples for Confirmation of Diagnosis
Treatment
Primary Treatment: Antimicrobial Therapy
Ruminants
Horses
Supportive Therapy
Control
Prevention of Introduction of Infection (Biosecurity)
Limitation of Spread Within a Herd
Principles of Infectious Disease Control for Prevention of Nosocomial Gastrointestinal and Respiratory Diseases in Large-Animal Hospitals
Reducing Exposure to Pathogens
Avoiding Increasing Susceptibility to Pathogens
Monitoring Effectiveness of the Infectious Disease Control Program
Recommended Steps in Developing an Effective Infectious Disease Control Program for a Large-Animal Hospital
Animals Being Transported
Immunization
Salmonella Vaccinology
Cattle
Horses
Sheep
Further Reading
References
Acute Undifferentiated Diarrhea of Newborn Farm Animals (Particularly Calves and Piglets)
Risk Factors
Animal Risk Factors
Colostrum
Environmental and Management Risk Factors
Calves
Nutrition of the Dam in the Preparturient Period
Calving Management: Dairy
Calving Management: Beef
Colostrum Management: Dairy
Calf Housing and Feeding: Dairy
Calf Housing and Feeding: Beef
Disease Control and Management: Dairy
Other Environmental and Management Factors
Piglets
Lambs
Pathogen Risk Factors
Calves
Lambs and Goat Kids
Piglets
Foals
Clinical Management of Epidemics
Control
Further Reading
References
Enterocolitis Associated With Clostridium Difficile
Etiology
Epidemiology
Occurrence
Horses
Pigs
Pathogen Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Horses
Pigs
Clinical Pathology
Necropsy Findings
Horses
Pigs
Treatment
Control
Further Reading
References
Proliferative Enteropathy in Horses
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Necropsy
Samples for Confirmation of Diagnosis
Treatment and Control
References
Equine Neorickettsiosis (Equine Monocytic Ehrlichiosis, Equine Ehrlichial Colitis, and Potomac Horse Fever)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
References
Equine Coronavirus Infection
References
Viral Diarrhea in Calves, Lambs, Kids, Piglets, and Foals
Etiology
Rotaviruses
Coronaviruses
Toroviruses
Parvovirus
Other Viruses and Mixed Infections
Epidemiology
Occurrence
Methods of Transmission
Immune Mechanisms
Calves: Bovine Rotavirus
Occurrence and Prevalence of Infection
Concurrent Infections
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogen Risk Factors
Morbidity and Case Fatality
Method of Transmission
Immune Mechanisms
Bovine Coronavirus (Calf Diarrhea)
Parvovirus
Bovine Torovirus (Breda Virus)
Bovine Norovirus
Lambs and Kids
Rotavirus
Piglets
Porcine Rotavirus
Porcine Coronavirus
Foals
Equine Rotavirus
Pathogenesis
Rotavirus
Coronavirus
Porcine Coronavirus
Calicivirus-Like (Norovirus) Agent
Parvovirus
Clinical Findings
Calves
Lambs
Piglets
Foals
Clinical Pathology
Detection of Virus
Electron Microscopy
Immunofluorescence
Immunodiffusion and Electron Microscopy
Reverse Passive Hemagglutination, ELISA, and Polyacrylamide Gel Electrophoresis
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Management and Colostral Intake
Vaccination
Oral Vaccines to Newborn Calves
Vaccination of Pregnant Dam (for Passive Immunization of the Neonate Through Colostral Immunoglobulin)
Experimental Studies of Maternal Bovine Rotavirus Vaccines
Bovine Coronavirus Vaccine
Commercial Bovine Rotavirus–Coronavirus and E. Coli F5 (K99) Vaccines
Stored Colostrum
Systemic Colostral Antibody
Porcine Rotavirus Vaccines
Equine Rotavirus Vaccine
Subunit Vaccines
Further Reading
References
Vesicular Stomatitis (Sore Mouth, Indiana Fever)
Etiology
Epidemiology
Occurrence
Geographic Occurrence
Host Occurrence
Morbidity and Mortality
Method of Transmission
Risk Factors
Host Risk Factors
Environmental Risk Factors
Pathogen Risk Factors
Experimental Reproduction
Economic Importance
Zoonotic Implications
Pathogenesis
Immune Mechanisms
Clinical Findings
Cattle
Horses
Pigs
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Parasitic Diseases of the Alimentary Tract
Cryptosporidiosis
Etiology
Epidemiology
Occurrence and Prevalence
Calves
Sheep and Goats
Pigs
Foals
Farmed Deer
Source of Infection and Transmission
Risk Factors
Pathogen Risk Factors
Concurrent Infections
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Prevention and Control
Treatment Options
Supportive Treatment
Management Strategies
Further Reading
References
Coccidiosis
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Calves
Adult Cattle
Sheep and Goats
Pigs
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Multiinfections
Immune Mechanisms
Pathogenesis
Life Cycle
Clinical Findings
Cattle and Sheep
Coccidiosis With Nervous Signs
Lambs
Piglets
Clinical Pathology
Fecal Oocyst Counts
Calves
Lambs
Piglets
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Calves and Lambs
Piglets
Control
Management of Environment
Lambs at Pasture
Feedlot Cattle and Lambs
Piglets
Coccidiostats
Antimicrobials
Ionophores
Vaccines
Further Reading
References
Giardiasis
Etiology
Epidemiology
Occurrence
Source of Infection
Pathogen Risk Factors
Animal and Management Risk Factors
Experimental Studies
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Ascariasis in Pigs, Horses, and Cattle
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Pigs
Horses
Buffalo Calves
Control
Further Reading
References
Strongylosis (Cyathostominosis) in Horses
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Oxyuris Equi (Pinworm)
Further Reading
References
Strongyloides (Threadworm)
References
Trichuris (Whipworm)
Chemotherapy
References
Parasitic Gastritis in Pigs
Etiology
Life Cycles
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
References
Gasterophilus spp. Infestation (Botfly)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnosis
Treatment
Control
Treatment Recommendations
Further Reading
References
Thorny-Headed Worm IN Pigs (Macracanthorhyncus hirudinaceus)
Tapeworm Infestations
Larval Tapeworm Infestation
Further Reading
Reference
Adult Tapeworm Infestation
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Toxins Affecting the Alimentary Tract
Phosphorus Toxicosis
Clinical Findings
Necropsy Findings
Diagnostic Confirmation
Samples for Analysis
Treatment
References
Arsenic Toxicosis
Etiology
Background Information
Relative Toxicities
Epidemiology
Occurrence
Dips and Sprays
Herbicides
Insecticidal Sprays
Leaded Gasoline
Insect Baits
Wood Preservatives
Metal-Bearing Ore Deposits
Pharmaceuticals and Growth Stimulants
Animal Risk Factors
Human Risk Factors
Environmental Risk Factors
Pathogenesis
Mechanism of Action
Tissue Susceptibility
Time Lag
Percutaneous Absorption
Chronic Poisoning
Nervous Tissue Lesions
Clinical Findings
Gastrointestinal Syndrome
Peracute Cases
Acute and Subacute Cases
Chronic Cases
Neurological Syndrome
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Toxicology:
Histology:
Treatment
Control
Further Reading
References
Molybdenum Toxicosis (Molybdenosis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Transmission
Pathogenesis
Clinical Findings
Acute Intoxication
Chronic Intoxication
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Amitraz Toxicosis
Etiology
Pathogenesis
Clinical Findings
Treatment
Further Reading
References
Propylene Glycol Toxicosis
Further Reading
Plant Materials Causing Physical Damage
Colic in horses from ingestion of indigestible fiber is associated with
Ruminal impaction in cattle is associated with ingestion of cuttings from
Further Reading
References
Plant Toxins Affecting the Alimentary Tract
Andromedotoxin
Anthraquinone
Colchicine
Irritant Diterpenoids
Irritant Oils
Lycorine
Podophyllin Poisoning
Protoanemonin Poisoning
Toxalbumins (Lectins)
Further Reading
References
Plants (Unidentified Toxins) Affecting the Gastrointestinal Tract
Diarrhea: Without Gastroenteritis as a Lesion
Diarrhea: With Gastroenteritis as a Lesion, Often With Abdominal Pain and Incoordination, Sometimes With Dysentery and Vomiting
Dysphagia
Esophageal Ulceration
Salivation With OR Without Stomatitis
Vomiting
Slaframine Toxicosis (Slobbers, Black Patch Disease)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Farm or Premise Risk Factors
Transmission
Pathogenesis
Clinical Findings
Horses
Ruminants
Swine
Necropsy Findings
Treatment
Control
Further Reading
References
Cantharidin Toxicosis (Blister Beetle Poisoning, Canthariasis)
Etiology
Epidemiology
Occurrence
Risk Factors
Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Neoplasms of the Alimentary Tract
Mouth
Pharynx and Esophagus
Stomach and Rumen
Intestines
References
Tumors of the Peritoneum
Reference
Congenital Defects of the Alimentary Tract
Harelip and Cleft Palate
Atresia of the Salivary Ducts
Agnathia, Micrognathia, and Brachygnathia
Persistence of the Right Aortic Arch
Choanal Atresia
Congenital Atresia of the Intestine and Anus
Intestinal Atresias
Atresia of the Terminal Colon
Atresia of the Anus
Multiple Organ Defects
Further Reading
References
Inherited Defects of the Alimentary Tract
Inherited Defects of the Mouth and Jaw
Cleft Palate
Jaw Deformity
Smooth Tongue (Epitheliogenesis Imperfecta Linguae Bovis)
Tongue Aplasia
Rectal Prolapse
Inherited Rectovaginal Constriction
Inherited Atresia of Alimentary Tract Segments
Lethal White Syndrome IN Foals and Lambs (Intestinal Aganglionosis)
References
8 Diseases of the Alimentary Tract–Ruminant
Diseases of the Forestomach of Ruminants
Anatomy and Physiology
Reticulorumen Motility
Primary Contraction Cycle
Control of Primary Contractions
Ruminal Atony
Hypomotility
Properties of Contractions
Extrinsic Control of Primary Contractions
Excitatory Inputs to the Gastric Center
Inhibitory Inputs to the Gastric Center
Fever
Endotoxemia
Pain
Distension of Forestomach
Ruminal Volatile Fatty Acids
Abomasal Disease
Effect of Depressant Drugs
Acid-Base Imbalance and Blood Glucose
Hormonal Control of Primary Contractions
Intrinsic Control of Primary Contractions
Treatment of Forestomach Hypomotility
Secondary Cycle Contraction and Eructation
Rumination
Esophageal Groove Closure
Ruminant Gastrointestinal Dysfunction
Further Reading
Special Examination of the Alimentary Tract and Abdomen of Cattle
History
Systemic State, Habitus, and Appetite
Oral Cavity and Esophagus
Inspection of the Abdomen
Distension of the Abdomen
Lavage of Distended Rumen
Left Side of Abdomen and Rumen
Inspection and Palpation
Nature of Rumen Contents
Auscultation of the Rumen and Left Flank
Auscultation of Rumen
Fluid-Tinkling or Fluid-Splashing Sounds
Right Side of Abdomen
Examination of Rumen Fluid
Analysis of Rumen Fluid
Palpation Per Rectum of the Abdomen
Gross Examination of Feces
Amount
Absence of or Scant Feces
Color
Odor
Consistency
Degree of Digestion
Other Substances in the Feces
Mucus
Fibrin
Detection of Abdominal Pain
Ultrasonography
Radiography
Endoscopy of the Rumen
Computed Tomography
Rumination Monitors
Serum Biomarkers of Gastrointestinal Function
Interpretation of Clinical Findings
Exploratory Laparotomy
Intussusception and Other Strangulation Obstructions of the Small Intestines
“Atypical” Left-Side Displacement of the Abomasum
Traumatic Reticuloperitonitis
Laparoscopy
Clinical Examination of the Digestive Tract and Abdomen of the Calf
Abdominal Distension in Calves
Further Reading
References
Diseases of the Rumen, Reticulum and Omasum
Simple Indigestion
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Spontaneous Recovery
Rumenatorics
Parasympathomimetics
Alkalinizing and Acidifying Agents
Reconstitution of Ruminal Microflora (Rumen Transfaunation)
Further Reading
References
Rumen Impaction Due to Indigestible Foreign Bodies
References
Indigestion in Calves Fed Milk Replacers (Ruminal Drinkers)
References
Acute Carbohydrate Engorgement of Ruminants (Ruminal Lactic Acidosis, Rumen Overload) and Subacute Ruminal Acidosis
Etiology
Epidemiology
Occurrence
Previous Diet and Change of Ration
Accidental Consumption of Excess Carbohydrates
Feedlot Cattle
Beef Breeding Herds
Lamb Feedlots and Liquid-Fed Calves
Dairy Cattle Herds
Morbidity and Case–Fatality Rates
Types and Toxic Amounts of Feeds
Pathogenesis
Changes in Rumen Microflora
Volatile Fatty Acids and Lactic Acid in the Rumen
Systemic Lactic Acidosis
Chemical and Mycotic Rumenitis
Hepatic Abscesses
Laminitis
Other Toxic Substances Produced
Experimental Lactic Acidosis
Subacute Ruminal Acidosis (Dairy Cattle)
Clinical Findings
Speed of Onset and Severity
Individual Animals
Ultrasonography
Mycotic Rumenitis
Complications
Subacute Ruminal Acidosis in Dairy Cattle
Clinical Pathology
Ruminal Fluid pH
Ruminal Protozoa
Serum Biochemistry
Fecal pH
Urine pH
Low Milk-Fat Percentage
Necropsy Findings
Treatment
Triage
Rumenotomy
Intravenous Sodium Bicarbonate and Fluid Therapy
Rumen Lavage
Intraruminal Alkalinizing Agents
Ruminal Transfaunation
Ancillary Therapy
Monitor Response to Therapy
Control and Prevention
Total Mixed Rations
Small Incremental Increases in Concentrate
Feedlot Starter Rations
Dietary Buffers
Ionophores
Subacute Ruminal Acidosis in Dairy Cattle
Limiting the Intake of Rapidly Fermentable Carbohydrates
Providing Adequate Ruminal Buffering
Allowing for Ruminal Adaptation to High-Grain Diets
Feeding Management in Early Lactation
Vaccination Against Lactic Acidosis
Further Reading
References
Ruminal Parakeratosis
Ruminal Tympany (Bloat)
Etiology
Primary Ruminal Tympany (Frothy Bloat)
Pasture and Feedlot Bloat
Frothy Rumen Contents
Role of Saliva
Secondary Ruminal Tympany (Free-Gas Bloat)
Chronic Ruminal Tympany
Epidemiology
Occurrence
Pasture Bloat
Feedlot Bloat
Morbidity and Case Fatality
Pasture Bloat
Feedlot Bloat
Risk Factors That Influence the Occurrence of Primary Ruminal Tympany
Dietary Risk Factors
Bloating Forages
Nonbloating Forages
Crop Maturity
Weather Risk Factors
Feedlot Bloat
Animal Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Primary Pasture or Feedlot Bloat
Secondary Bloat
Dyspnea and Tachycardia in Severe Bloat
Clinical Pathology
Necropsy Findings
Treatment
First-Aid Emergency Measures
Emergency Rumenotomy
Trocar and Cannula
Promote Salivation
Stomach Tube
Feedlot Bloat
Antifoaming Agents
Return to Pasture or Feed
Control
Pasture Bloat
Management Strategies to Reduce Rate of Rumen Fermentation
Stage of Growth
Choice of Forages
Alternative Temperate Forages
Grazing Management
Grazing Patterns and Strip Grazing
Swathing and Wilting
Alfalfa Hay Bloat Prevention
Antifoaming Agents
Oils and Fats
Individual Drenching
Application of Oil to Pasture
Addition to Feed and Water
Application to Flanks
Types of Oil
Water-Soluble Feed Supplements
Synthetic Nonionic Surfactants
Polyoxythylene-Polyoxypropylene Block Polymer
Polyoxypropylene-Polyoxyethylene Glycol Surfactant Polymer (Alfasure)
Alcohol Ethoxylate Detergents
Ionophores
Controlled-Release Monensin Capsules
Liquid Formulation of Monensin
Feedlot Bloat
Roughage in Ration
Consistency of Grain
Antifoaming Agents
Dietary Salt
Genetic Control of Pasture Bloat
General Comments
Further Reading
Reference
Traumatic Reticuloperitonitis
Etiology
Epidemiology
Occurrence
Risk Factors
Economic Importance
Pathogenesis
Ingestion of Foreign Body
Penetration of Reticulum
Acute Local Peritonitis
Generalized Peritonitis and Extension of Disease
Clinical Findings
Acute Local Peritonitis
Chronic Local Peritonitis
Rectal Examination
Acute Diffuse (Generalized) Peritonitis
Sudden Death
Iatrogenic Reticulitis
Clinical Pathology
Hemogram
Plasma Protein, Fibrinogen, Acute Phase Reactants, and Coagulation Profile
Abdominocentesis and Peritoneal Fluid
Radiography of Cranial Abdomen and Reticulum
Ultrasonography of the Reticulum
Ultrasonography for Traumatic Reticuloperitonitis
Ultrasonography and Radiography of Cattle With Traumatic Reticuloperitonitis
Metal Detection
Laparoscopy
Necropsy Findings
Treatment
Conservative Medical Therapy
Antimicrobials
Rumenotomy
Drainage of Reticular Abscesses
Choice of Treatment
Prevention
Reticular Magnets
Further Reading
References
Vagus Indigestion
Etiology
Complications of Traumatic Reticuloperitonitis
Vagal Nerve Injury and Dysfunction
Reticular Adhesions
Other Causes
Epidemiology
Pathogenesis
Anterior Functional Stenosis (Achalasia)
Posterior Functional Stenosis (Achalasia)
Metabolic Alkalosis and Abomasal Reflux
Postsurgical Complication in Abomasal Volvulus
Abomasal Impaction in Sheep
Clinical Findings
Ruminal Distension With Hypermotility
Ruminal Distension With Atony
Pyloric Obstruction and Abomasal Impaction
Clinical Pathology
Hemogram
Peritoneal Fluid
Serum Biochemistry
Ruminal Chloride Concentrations
Necropsy Findings
Treatment
Rumen Lavage
Fluid and Electrolyte Therapy and Laxatives
Rumenotomy
Prevention
Further Reading
References
Diaphragmatic Hernia
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Traumatic Reticulopericarditis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Hemogram
Pericardiocentesis
Necropsy Findings
Treatment
Prevention
Further Reading
Reference
Traumatic Reticulosplenitis and Reticulohepatitis
Impaction of the Omasum
References
Diseases of the Abomasum
Clinical Examination of the Abomasum
Physical Examination
Ultrasonography of the Abomasum
Abomasocentesis
Applied Anatomy and Pathophysiology of the Abomasum
Abomasal Reflux
Further Reading
References
Left-Side Displacement of the Abomasum
Etiology
Epidemiology
Occurrence
Calves
Lactational Incidence Rate
Case Fatality
Risk Factors
Dietary Risk Factors
Prepartum Nutrition and Management
High-Level Grain Feeding
Dietary Crude Fiber
Animal Risk Factors
Breed and Age of Cow
Season of the Year
Influence of Weather
Milk Production
Late Pregnancy
Concurrent Diseases
Preexisting Subclinical Ketosis and Hepatic Lipidosis
Hypocalcemia
Metabolic Predictors of Left-Side Displacement of Abomasum
Genetic Predisposition
Miscellaneous Animal Risk Factors
Economic Importance and Effects on Production and Survivorship
Pathogenesis
Abomasal Luminal Gas Pressure, Volume, and Perfusion in Cows With Left Displaced Abomasum or Abomasal Volvulus
Perforating Abomasal Ulceration
Clinical Findings
General Appearance and Ketosis
Status of Reticulorumen and Spontaneous Abomasal Sounds
Pings of the Left Displaced Abomasum
Acute Left Displaced Abomasum
Concurrent Perforating Abomasal Ulceration
Other Clinical Features
Ultrasound Examination
Rectal Examination
Secondary Ketosis and Hepatic Lipidosis
Atrial Fibrillation
Course of Left-Side Displacement of the Abomasum
Unusual Cases of Left-Side Displacement
Left-Side Displacement of the Abomasum in Calves
Clinical Pathology
Hemogram
Serum Biochemistry
Cowside Tests of Blood β-Hydroxybutyrate Concentration and Urinary Ketones
Peritoneal Fluid Analysis
Abomasocentesis
Necropsy Findings
Treatment
Open Surgical Techniques
Laparoscopic Techniques
Closed Suture Techniques
Prokinetic and Analgesic Administration
Survivorship Following Surgery to Correct Left-Side Displacement of the Abomasum
Treatment of Ketosis
Rumen Transfaunation Following Surgery for Left-Side Displacement of the Abomasum
Control
Prepartum Nutrition and Management
Crude Fiber Intake
Monensin in Controlled-Release Capsule Prepartum
Genetic Selection
Further Reading
References
Right-Side Displacement of the Abomasum and Abomasal Volvulus
Etiology
Epidemiology
Occurrence and Incidence
Lactating Dairy Cows
Beef Cattle
Calves
Mature Bulls and Pregnant Cows
Risk Factors
Pathogenesis
Definition of Right Displaced Abomasum and Abomasal Volvulus
Dilatation and Displacement Phase
Volvulus Phase
Postsurgical Complication in Right-Side Displacement of the Abomasum or Abomasal Volvulus
Clinical Findings
Right Displaced Abomasum and Abomasal Volvulus (Adult Cattle)
Acute Abomasal Volvulus (Calves)
Postsurgical Complication in Abomasal Volvulus
Clinical Pathology
Serum Biochemistry
Urinalysis
Hemogram
Ultrasonography of Right Abdomen
Peritoneal Fluid Analysis
Abomasocentesis
Prognostic Indicators
Necropsy Findings
Treatment
Surgical Correction
Prokinetic Administration
Fluid and Electrolyte Therapy
Oral Therapy
Deflation of Distended Abomasum in Calves
Control
Further Reading
References
Abomasal Impaction in Cattle
Etiology and Epidemiology
Primary Abomasal Impaction
Dietary Abomasal Impaction
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Surgery
Control
Primary Abomasal Impaction
Dietary Abomasal Impaction
References
Abomasal Impaction in Sheep and Goats (Abomasal Emptying Defect)
Reference
Abomasal Phytobezoars and Trichobezoars
Reference
Abomasal Ulcers of Cattle
Etiology
Primary Ulceration
Secondary Ulceration
Epidemiology
Primary Abomasal Ulcers
Lactating Dairy Cows
Mature Bulls and Feedlot Cattle
Hand-Fed Calves
Veal Calves
Sucking Beef Calves
Dietary Factors in Calves Fed Milk or Milk Replacer
Secondary Abomasal Ulcers
Pathogenesis
Type 1: Nonperforating Ulcer
Type 2: Ulcer Causing Severe Blood Loss
Type 3: Perforating Ulcer With Acute, Local Peritonitis
Type 4: Perforating Ulcer With Diffuse Peritonitis
Clinical Findings
Perforation of Ulcer
Nursing Beef Calves
Clinical Pathology
Melena
Hemogram
Plasma Gastrin and Pepsinogen Concentration
Microbiology
Necropsy Findings
Treatment
Blood Transfusions
Coagulants
Antacids
Histamine Type-2 Receptor Antagonists
Alkalinizing Agents
Kaolin and Pectin
Surgical Excision
Prevention
Further Reading
References
Omental Bursitis
Reference
Abomasal Bloat (Tympany) in Calves And Lambs
Further Reading
References
Diseases of the Intestines of Ruminants
Small and Large Intestinal Obstruction in Cattle
Etiology and Epidemiology
Intestinal Accidents
Volvulus
Intussusception
Strangulation
Compression Stenosis
Cecal Dilatation
Luminal Blockages
External Pressure
Ileal Impaction in Cows
Fiber Balls or Phytobezoars
Trichobezoars (Hairballs)
“Rectal Paralysis”
Duodenal Ileus
Functional Obstructions
Pathogenesis
Physical Obstruction
Volvulus and Intussusception
Incarceration
Duodenal Ileus
Functional Obstruction
Clinical Findings
General Findings
Abdomen
Feces
Palpation Per Rectum
Duodenal Ileus
Volvulus of the Spiral Colon (Mesenteric Root Volvulus)
Lipomas and Fat Necrosis
Clinical Pathology
Serum Biochemistry
Hemogram
Necropsy Findings
Treatment
Surgical Correction
Fluid Therapy
Antimicrobials
Nonsteroidal Antiinflammatory Drugs
Further Reading
References
Intestinal Obstruction in Sheep
Terminal Ileitis of Lambs
Cecal Dilatation and Cecocolic Volvulus in Cattle
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Ultrasonographic Examination of the Cecum
Clinical Pathology
Treatment
Medical Therapy
Surgical Correction
Further Reading
References
Bacterial Diseases of the Ruminant Alimentary Tract
Actinomycosis (Lumpy Jaw)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Cattle
Pigs
Clinical Pathology
Necropsy Findings
Treatment and Control
Reference
Actinobacillosis (Wooden Tongue)
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Cattle
Sheep
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Oral and Laryngeal Necrobacillosis
Etiology
Epidemiology
Occurrence
Transmission
Risk Factors
Host Risk Factors
Pathogen Risk Factors
Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Enterohemorrhagic Escherichia coli in Farm Animals and Zoonotic Implications
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Cattle
Prevalence of Infection in Cattle, Sheep, and Pigs at Slaughter
Sheep and Goats
Wildlife
Pigs
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Housing and Management Practices
Pathogen Risk Factors
Virulence Attributes and Mechanisms
Acid Resistance
Antimicrobial Resistance
Methods of Transmission
Sources of Organism
Ruminants as Reservoirs
Other Species
Wild Birds
Flies
Environmental Sources
Water Supplies for Livestock
Feed Supplies
Manure
Soil
Animal-Holding Facilities
Immune Mechanisms
Zoonotic Implications
Economic Importance
Pathogenesis
Experimental Reproduction
Clinical Pathology
Control
Specific Strategies for Control of Escherichia coli O157:H7 at Preharvest Level
Animal Management Strategies
Water Systems and Runoff
Environmental Control of STEC
Diet Changes
Direct Antipathogen Strategies
Vaccination Against Escherichia coli O157:H7
Competitive Enhancement Strategies
Probiotics
Sodium Chlorate Supplementation
Control of Escherichia coli O157:H7 During Slaughtering and Postharvest Stage
Meat Inspection Service and Surveillance
Postharvest Decontamination Techniques
Progress Made With Decontamination Processes
Irradiation
Consumer Education on Handling and Cooking Meat
Visitors to Animal Farms
Further Reading
References
Braxy (Bradsot)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
Enteric Disease Associated With Clostridium Perfringens
References
Enterotoxemia Associated With Clostridium Perfringens Type A
Enterocolitis in Horses
Equine Intestinal Clostridiosis
Enteritis in Piglets
Hemorrhagic Enterotoxemia and Hemolytic Disease in Cattle, Sheep, and Goats
Abomasal Ulcer
Jejunal Hemorrhage Syndrome in Cattle
Further Reading
References
Enterotoxemia Associated With Clostridium Perfringens Types B, C, and E
Etiology
Epidemiology
Lamb Dysentery and Type C Enterotoxemia
Occurrence
Animal and Environmental Risk Factors
Necrotic Enteritis of Pigs
Occurrence
Animal and Environmental Risk Factors
Enterotoxemia in Foals
Enterotoxemia in Calves
Struck in Sheep
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Outbreaks
Long-Term Control
Further Reading
References
Jejunal Hemorrhage Syndrome (Hemorrhagic Bowel Syndrome, Hemorrhagic Jejunitis, or Jejunal Hematoma) in Cattle
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Hematology
Serum Biochemistry
Necropsy Findings
Treatment
Control
Further Reading
References
Paratuberculosis (Johne’s Disease): Cattle
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Wildlife and Exotic Species
Prevalence of Infection
Methods of Transmission
Risk Factors
Animal Risk Factors
Age of Animal
Breed Incidence and Genetic Susceptibility
Other Diseases and Stressors
Herd Characteristics
Environmental and Management Risk Factors
Care of the Newborn Calf
Calf Rearing
Soil Characteristics and Manure Handling
Pathogen Risk Factors
Survival and Dormancy of Organism in the Environment
Thermal Resistance of Organism
Economic Importance
Losses at National Dairy Industry Level
Zoonotic Implications
Pathogenesis
Susceptibility to Infection
Immune Response and Spectrum
Clinical Findings
Stages of Disease
Stage One
Silent Infection.
Stage Two
Subclinical Disease.
Stage Three
Clinical Disease.
Stage Four
Advanced Clinical Disease.
Clinical Pathology
Diagnosis of MAP Infection
Culture or Detection of Organism
Bacteriologic Culture.
Pooled Fecal Samples and Culture.
DNA Probes and Polymerase Chain Reaction.
Culture of Milk and Blood.
Tests on Tissue Samples.
Serologic Tests
Complement Fixation Test.
Agar Gel Immunodiffusion.
Enzyme-Linked Immunosorbent Assay.
Immunity Tests.
Summary of Diagnostic Testing
Diagnostic Strategies for Different Situations
Necropsy Findings
Treatment
Control
Principles of Control
Identification and Elimination of Infected Animals
Prevention of Introduction of Infected Animals Into the Herd
Prevention of Exposure of Susceptible Animals to the Infectious Agent
Dairy Herds
Hygiene
Dairy Calf Health Management
Beef Herds
Vaccination of Cattle
Control on a Countrywide Basis
Johne’s Disease Control in the United States
Education of the Producer
Risk Assessment and Disease Management Plan
Herd Testing and Herd Classification
Johne’s Disease Control in the Netherlands
Further Reading
References
Paratuberculosis (Johne’s Disease): Sheep, Goats, Cervids, and Camelids
Etiology
Epidemiology
Occurrence, Morbidity, and Mortality
Sheep and Goats
Deer, Camelids, and Exotic Species
Prevalence and Source of Infection
Sheep
Methods of Transmission
Sheep
Deer
Risk Factors
Sheep and Management
Deer
Environmental Risk Factors
Soil Characteristics
Pathogen Risk Factors
Survival and Dormancy of Organism in the Environment
Zoonotic Implications
Pathogenesis
Immune Response
Development of Lesions
Clinical Findings
Sheep and Goats
Other Species (Deer, Camelids, and Bison)
Clinical Pathology
Diagnostic Tests
Culture or Detection of Organism
Bacteriologic Examination.
Pooled Fecal Samples and Culture.
Genetic Probe.
Biopsy.
Serologic Tests
Tests of Immunity
Serum Biochemistry
Deer
Necropsy Findings
Sheep and Goats
Samples for Confirmation of Diagnosis
Rabbits
Treatment
Control
Control on a Flock Basis
Vaccination
Goats
Control on a Countrywide Basis
Further Reading
References
Viral Diseases of the Ruminant Alimentary Tract
Rinderpest (Cattle Plague)
Synopsis of the Disease
Steps Leading to Eradication
Lessons From the Eradication
Further Reading
References
Peste Des Petits Ruminants (Goat Plague, or Kata)
Etiology
Epidemiology
Occurrence
Morbidity and Case–Fatality Rate
Methods of Transmission
Risk Factors and Immune Mechanisms
Experimental Reproduction
Economic Importance
Zoonotic Implication
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Bovine Viral Diarrhea, Mucosal Disease, and Bovine Pestivirus Disease Complex
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Other Ruminant Species
Morbidity and Case–Fatality Rates
Methods of Transmission
Direct Contact
Indirect Contact
Airborne Transmission
Flies
Fomites
Artificial Insemination/Embryo Transfer
Vaccines Contaminated With Bovine Viral Diarrhea Virus
Risk Factors
Host Risk Factors
Environmental and Management Risk Factors
Pathogen Risk Factors
Immune Mechanisms
Economic Importance
Pathogenesis
Immunocompetent Nonpregnant Cattle Subclinical Bovine Viral Diarrhea
Peracute Bovine Viral Diarrhea
Thrombocytopenia and Hemorrhagic Syndrome
Diarrhea of Neonatal Calves
Meningoencephalitis
Immunosuppression
In Vivo Evidence of Immunosuppression.
Bovine Viral Diarrhea Virus in the Feedlot
Ovarian Dysfunction
Immunocompetent Pregnant Cattle and Fetal Infections
Immunotolerant Persistently Infected Cattle
Clinical Findings
Inapparent or Subclinical Infection (Bovine Virus Diarrhea)
Acute Mucosal Disease
Chronic Mucosal Disease
Unthrifty Persistently Infected Calves
Peracute Bovine Virus Diarrhea
Thrombocytopenia and Hemorrhagic Disease
Reproductive Failure and Neonatal Disease
Congenital Defects in Calves
Clinical Pathology
Virus Isolation
Antigen Detection
Immunofluorescence or Immunohistochemistry.
Skin Biopsy.
Polymerase Chain Reaction Amplification
Serology
Serum Neutralization Test
Antibody Enzyme-Linked Immunosorbent Assay
Use of Laboratory Tests in the Herd
Acute Infections
Persistently Infected Animals
Prenatal Diagnosis of Persistent Infection
Herd Screening
Necropsy Findings
Acute Mucosal Disease
Peracute Bovine Viral Diarrhea
Chronic Mucosal Disease
Abortion
Samples for Confirmation of Diagnosis
Treatment
Control and Prevention
Identification and Elimination of Persistently Infected Animals From the Herd
Prevention of Introduction of Infection Into Herd (Biosecurity)
Immunization Programs and Biocontainment
Bovine Viral Diarrhea Virus Vaccines
Modified-Live Virus-Bovine Viral Diarrhea Virus Vaccines
Inactivated Bovine Viral Diarrhea Virus Vaccines
Combination Vaccines
Efficacy of Bovine Viral Diarrhea Virus Vaccines
Commercially Available Vaccines
Strategies for Bovine Viral Diarrhea Virus Vaccination Programs
Prevention of Fetal Infection in Dairy and Beef Herds
Postnatal Bovine Viral Diarrhea Virus Infections
Vaccination Schedules
Beef Cow–Calf Herd
Beef Feedlot
Dairy Herd
Current Vaccination Practices
Eradication of Bovine Viral Diarrhea Virus Infection Without Vaccination
Bovine Viral Diarrhea Control Programs in Different Geographic Regions and Countries
Scandinavian Countries
United Kingdom
Continental Europe
Further Reading
References
Winter Dysentery of Cattle
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Morbidity and Case–Fatality Rates
Methods of Transmission
Experimental Reproduction
Risk Factors
Host and Environmental Risk Factors
Pathogen Risk Factors
Pathogenesis
Clinical Findings
Cattle
Clinical Pathology
Detection of Virus
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Vaccination
Further Reading
References
Bovine Papular Stomatitis
Etiology
Epidemiology
Transmission
Clinical Findings
Clinical Pathology
Treatment
Control
Further Reading
References
Parasitic Diseases of the Ruminant Alimentary Tract
Parasitic Gastroenteritis in Ruminants
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Cattle
Sheep
Goats
Choice of Anthelmintic
Control
Further Reading
References
Hemonchosis in Ruminants
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Bunostomosis (Hookworm Disease) in Ruminants
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
References
Oesophagostomosis (Nodule Worm Disease) in Ruminants and Pigs
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Chabertiosis
References
Stomach Fluke Disease (Intestinal Amphistomosis)
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Toxic Diseases of the Ruminant Alimentary Tract
Nonprotein Nitrogen Toxicosis (Urea Toxicosis)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Pathogenesis
Clinical Findings
Cattle and Sheep
Clinical Pathology
Cattle
Sheep
Necropsy Findings
Treatment
Control
Further Reading
References
Chemically Treated Natural Feeds
Formalin-Treated Grain
Caustic-Treated Grain
Ammoniated Forage
Newsprint
Sewage Sludge
Further Reading
Ammoniated feeds:
Caustics:
Newsprint:
Sewage sludge:
Reference
Diseases of the Ruminant Alimentary Tract of Unknown Cause
Chronic Inflammatory Bowel Disease of Sheep
Further Reading
Salivary Abomasum Disease
References
9 Diseases of the Liver
Diseases of the Liver: Introduction
Principles of Hepatic Dysfunction
Diffuse and Focal Hepatic Disease
Hepatic Dysfunction
Portal Circulation
Manifestations of Liver and Biliary Disease
Jaundice (Icterus)
Prehepatic or Hemolytic Jaundice
Hepatic or Hepatocellular Jaundice
Posthepatic Jaundice
Clinical Findings
Clinical Pathology
Hepatic Encephalopathy
Edema and Emaciation
Diarrhea and Constipation
Photosensitization
Hemorrhagic Diathesis
Abdominal Pain
Alteration in Size of the Liver
Displacement of the Liver
Rupture of the Liver
Black Livers of Sheep
References
Special Examination of the Liver
Palpation and Percussion
Biopsy
Medical Imaging of the Liver
Ultrasonography
Radiography
Laboratory Tests for Hepatic Disease and Function
Hepatic Function Tests
Serum Bile Acids
Serum Bilirubin
Ammonia
Urea Nitrogen
Albumin
Serum Hepatic Enzyme Tests
Hepatic Enzyme Profile According to Species
Cattle
Calves
Horses
Further Reading
References
Principles of Treatment in Diseases of the Liver
Diffuse Diseases of the Liver
Hepatitis
Etiology and Epidemiology
Toxic Hepatitis
Poisonous Plants, Fungi, and Insects
Miscellaneous Farm Chemicals
Toxemia Perfusion Hepatitis
Infectious Hepatitis
Parasitic Hepatitis
Nutritional Hepatitis (Trophopathic Hepatitis)
Idiopathic Hepatosis and Cirrhosis
Congestive Hepatopathy
Portosystemic Vascular Anomaly
Pathogenesis
Liver Disease and Liver Failure
Intravascular Hemolysis in Equine Liver Disease
Clinical Findings
Portosystemic Shunts
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
Liver Abscess and Necrobacillosis of the Liver
Etiology
Epidemiology
Occurrence
Pathogen Risk Factors
Risk Factors in Grain-Fed Cattle
Management
Diet
Breed
Risk Factors in Other Farm Animals
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Feed Management
Prophylactic Antimicrobial Therapy
Vaccination
Control in Young Lambs
Further Reading
References
Bacillary Hemoglobinuria (Red Water Disease)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Infectious Necrotic Hepatitis (Black Disease)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Source of Infection
Other Species
Pathogenesis
Clinical Findings
Sheep
Cattle
Horses
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
Clostridium novyi Infection
Etiology
Pathogenesis and Epidemiology
Clinical Signs
Pathology
Diagnosis
Treatment
Prevention
Reference
Diseases Characterized by Systemic Involvement
Acute Hepatitis (Postvaccinal Hepatitis) of Horses (Theiler’s Disease, Serum Hepatitis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Hepatitis E Virus (HEV)
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Treatment and Control
References
Hepatic Diseases Associated With Trematodes
Fasciolosis (Liver Fluke Disease)
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Acute Fasciolosis
Subacute Fasciolosis
Chronic Fasciolosis
Clinical Pathology
Necropsy Findings
Acute Hepatic Fasciolosis
Chronic Hepatic Fasciolosis
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Fascioloides magna
Further Reading
References
Dicrocoelium
Further Reading
References
Diseases Associated With Major Phytotoxins
Pyrrolizidine Alkaloid Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Plant Factors
Human Risk Factors
Transmission
Pathogenesis
Hepatic Injury
Hepatic Encephalopathy
Toxemic Jaundice
Secondary Photosensitization
Clinical Findings
Cattle
Horses
Pigs
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Plants Causing Hepatic Injury (Unidentified Toxin)
Hepatic Injury-Dummy Syndrome (Circling, Head Pressing, Compulsive Walking, and Blindness)
Hepatic Injury: Jaundice and/or Photosensitization
Poisoning by Mycotoxins
Poisoning by Aflatoxins (Aflatoxicosis)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Cattle
Pigs
Horses
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Phomopsins Toxicosis (Lupinosis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Lesions
Treatment
Control
Further Reading
References
Pithomycotoxicosis (Sporidesmin Toxicity and Facial Eczema)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Factors
Plant Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Rubratoxin Toxicosis
Further Reading
Reference
Miscellaneous Fungi Causing Hepatic Damage (Unidentified Toxin)
Sawfly Larvae (Lophyrtomin and Pergidin) Toxicosis
Etiology
Epidemiology
Clinical Signs
Necropsy Lesions
Further Reading
References
Coal Tar Pitch Poisoning in Pigs
Etiology
Clinical Findings
Necropsy Findings
Focal Diseases of the Liver
Tumors of the Liver
Diseases of the Biliary System
Cholangiohepatitis in Horses
Diseases of the Pancreas
Pancreatitis
Diabetes Mellitus
Pancreatic Adenoma
Pancreatic Adenocarcinoma
10 Diseases of the Cardiovascular System
Principles of Circulatory Failure
Heart Failure
Circuit Failure
Cardiac Reserve and Compensatory Mechanisms in Heart Failure
Cardiac Reserve and Heart Rate
Cardiac Reserve and Stroke Volume
Cardiac Reserve and Mixed Venous Oxygen Tension
Cardiac Reserve and Autonomic Nerve Activity
Cardiac Reserve in Cardiac Insufficiency
Measurement of Cardiac Reserve
Cardiac Enlargement
References
Manifestations of Circulatory Failure
Chronic (Congestive) Heart Failure
Etiology
Valvular Disease
Myocardial Disease
Congenital Anatomic Defects Producing Shunts
Hypertension
Pressure Load
Volume Load
Pumping Defects (Systolic Failure)
Filling Defects (Diastolic Failure)
Pathogenesis
Right-Sided Congestive Heart Failure
Left-Sided Congestive Heart Failure
Clinical Findings
Right-Sided Congestive Heart Failure
Left-Sided Congestive Heart Failure
Clinical Pathology
Necropsy Findings
Treatment
Diuretics
Angiotensin-Converting Enzyme Inhibitors
Stall Rest
Positive Inotropic Agents (Dobutamine, Calcium, Cardiac Glycosides)
Further Reading
References
Acute Heart Failure
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Special Examination of the Cardiovascular System
Physical Examination
Heart Sounds
First Heart Sound
Second Heart Sound
Third Heart Sound
Fourth Heart Sound
Sequence of Heart Sounds
Variation in Heart Sound Intensity
Heart Rate
Examination of the Arterial Pulse
Pulse Rate
Pulse Rhythm
Amplitude
Examination of the Jugular Vein
Percussion of the Thorax to Identify the Cardiac Dullness Area
Measurement of Jugular Venous Pressure
References
Measurement of Central Venous Pressure
References
Exercise Tolerance
Further Reading
Electrocardiography
Electrocardiograph
Depolarization and Repolarization
Lead Systems
Telemetry
Fetal Electrocardiography
Heart Rate Variability
Other Uses of the Electrocardiogram
Further Reading
References
Biomarkers of Myocardial Injury
Further Reading
References
Measurement of Arterial Blood Pressure
Measurement of Pulmonary Artery Blood Pressure
References
Echocardiography
Fetal Echocardiography
Further Reading
References
Radiographic and Angiocardiographic Examination
References
Phonocardiography
Reference
Cardiac Output
References
Cardiac Catheterization
Arrhythmias (Dysrhythmias)
Further Reading
References
Sinus Tachycardia, Sinus Bradycardia, and Physiologic Dysrhythmias
Sinus Tachycardia
Sinus Bradycardia
Physiologic Dysrhythmias
Reference
Arrhythmias With Normal Heart Rates or Bradycardia
Sinus Arrhythmia
Sinoatrial Block
Atrioventricular Block
First-Degree Atrioventricular Block
Second-Degree Atrioventricular Block
Third-Degree or Complete Heart Block
Premature Complexes
Atrial Premature Complexes
Junctional Premature Complexes
Ventricular Premature Complexes
References
Arrhythmias With Tachycardia
Atrial Fibrillation
Atrial Fibrillation in the Horse
Lone Atrial Fibrillation
Secondary Atrial Fibrillation
Atrial Fibrillation in the Cow
Atrial Fibrillation in the Sheep and Goat
Treatment of Atrial Fibrillation
Horses
Ruminants
Paroxysmal Tachycardia
Ventricular Tachycardia
Treatment
Ventricular Fibrillation
Further Reading
References
Diseases of the Heart
Valvular Disease and Murmurs
Etiology
Acquired
Congenital
Epidemiology
Horses
Cattle
Pigs
Pathogenesis
Generation of Murmurs
Valve Lesions
Murmurs Without Valvular Disease
Functional Murmurs
Effects of Valvular Disease
Cardiac Reserve
Clinical Findings
Technique of Examination
Timing
Duration
Intensity
Location and Radiation
Character
Interpretation
Functional (Innocent) Murmurs
Insufficiency of the Right AV Valve
Insufficiency of the Left Atrioventricular Valve
Insufficiency of the Aortic Valve
Stenosis of the Aortic Valve
Stenosis and Insufficiency of the Pulmonary Valve
Stenosis of the Right or Left Atrioventricular Valves
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Endocarditis
Etiology
Cattle
Horses
Pigs and Sheep
Epidemiology
Pathogenesis
Valve Predilection
Clinical Findings
Cardiac Signs
Embolism
Clinical Course
Rupture of the Chordae Tendineae
Electrocardiography
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Myocardial Disease and Cardiomyopathy
Etiology
Bacterial Myocarditis
Viral Myocarditis
Parasitic Myocarditis
Nutritional Deficiency
Toxicity
Venoms
Embolic Infarction
Tumor or Infiltration
Inherited
Unknown or Uncertain Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Rupture of the Heart and Acute Cardiovascular Accidents
Rupture of the Aorta
Rupture of Heart Valves
References
Cor Pulmonale
References
High Altitude Pulmonary Hypertension (Brisket Disease, Mountain Sickness)
Etiology
Epidemiology
Occurrence
Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Diagnostic Confirmation
Treatment
Control
Further Reading
References
Encephalomyocarditis Virus Disease in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation
Diagnosis
Treatment and Control
References
Cardiac Toxicities
Cardiac Glycoside Toxicosis
Etiology
Acute Poisoning
Chronic Poisoning: Cotyledonosis or Krimpsiekte
Epidemiology
Occurrence
Risk Factors
Animal Factors
Plant Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Acute Poisoning
Chronic Poisoning
Clinical Pathology
Necropsy Findings
Treatment
Primary Treatment
Supportive Treatment
Control
Further Reading
References
Yew (Taxus spp.) Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Transmission
Pathogenesis
Clinical Findings
Necropsy Findings
Treatment
Control
Further Reading
References
Monofluoroacetate Plant Toxicosis
References
4-Methoxypyridone Plant Toxicosis
Reference
Plants Causing Heart Failure (Unidentified Toxins)
Reference
Ionophore (Carboxylic) Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Swine Risk Factor (Tiamulin)
Farm Risk Factors
Pathogenesis
Clinical Signs
Cattle
Sheep
Swine
Horses
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Cardiac Neoplasia
References
Congenital Cardiovascular Defects
Etiology
Epidemiology
Cattle
Sheep
Horses
Pigs
Pathogenesis
Shunts
Age at Manifestation
Clinical Findings
Ectopic Heart
Patent Foramen Ovale
Ventricular Septal Defects
Complications
Prognosis
Tetralogy of Fallot
Patent Ductus Arteriosus
Complete Transposition of the Great Arteries
Persistent Truncus Arteriosus
Coarctation of the Aorta
Double-Outlet Right Ventricle
Fibroelastosis
Subvalvular Aortic Stenosis
Parachute Left Atrioventricular Valve
Valvular Abnormalities
Anomalous Origin of Coronary Arteries
Persistence of the Right Aortic Arch
Aberrant Right Subclavian Artery
Further Reading
References
Inherited Cardiovascular Defects
Bovine Hereditary Dilated Cardiomyopathy
Type 1 Calf: Acute Heart Failure
Type 2 Calf: Pulmonary Edema
Type 3: Young Adult Congestive Heart Failure
Reference
Inherited Ventricular Septal Defect
Inherited Aortic Aneurysm
Bovine Marfan Syndrome
Reference
Diseases of the Pericardium
Pericarditis
Etiology
Cattle
Horses
Sheep and Goats
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Diseases of the Blood Vessels
Arterial Thrombosis, Embolism, and Rupture
Etiology
Coagulopathies
Parasitic Arteritis
Viral Arteritis
Bacterial Arteritis
Embolic Arteritis and Thromboembolism
Microangiopathy
Calcification
Vasoconstrictive Agents
Epidemiology
Pathogenesis
Clinical Findings
Aortoiliac Thrombosis in the Horse
Aortoiliac Thrombosis in Calves
Pulmonary Embolism
Rupture of Abdominal Artery Aneurysm in Holstein-Friesian Cattle
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Purpura Hemorrhagica
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Venous Thrombosis
Thrombophlebitis
Clinical Signs
Treatment
Further Reading
References
Elaeophoriasis (Filarial Dermatitis in Sheep)
References
Vascular Neoplasia
Hemangioma and Hemangiosarcoma
Hemangioma
Hemangiosarcoma
Reference
11 Diseases of the Hemolymphatic and Immune Systems
Abnormalities of Plasma Protein Concentration
Hypoproteinemia
Etiology
Panhyproteinemia
Hypoalbuminemia
Hypogammaglobulinemia
Hypofibrinogenemia
Pathophysiology
Clinical Signs
Clinical Pathology
Necropsy
Treatment
Hyperproteinemia
Etiology
Panhyperproteinemia
Hyperglobulinemia
Hyperfibrinogenemia
Pathophysiology
Clinical Signs
Clinical Pathology
Necropsy
Treatment
References
Hemorrhagic Disease
Diagnosis
Treatment of Coagulopathies
Diseases Causing Hemorrhage
Vasculitis
Septicemic and Viremic Diseases
Purpura Hemorrhagica
Necrotizing Vasculitis
Treatment
Coagulation Defects
Acquired Hemostatic Defects
Inherited or Congenital Defects in Hemostasis
Hemophilia A
Von Willebrand Disease (Factor VIII : vWF Deficiency)
Factor XI Deficiency
Other Clotting-Factor Disorders
Platelet Disorders
Thrombocytopenia
Decreased Production
Increased Destruction
Inflammation and Infection.
Immune-Mediated (Idiopathic) Thrombocytopenia.
Increased Consumption
Other Causes
Thrombasthenia
Thrombocytosis
References
Disseminated Intravascular Coagulation and Hypercoagulable States
Etiology and Epidemiology
Prognosis
Pathophysiology
Clinical Signs
Clinical Pathology
Necropsy Examination
Diagnostic Confirmation
Treatment
References
Thrombosis (Hypercoagulability)
References
Diseases Characterized by Abnormalities of the Cellular Elements of the Blood
Disorders of Red Cell Number or Function
Anemia
Etiology
Hemorrhagic Anemia
Hemolytic Anemia
Cattle and Sheep
Pigs
Horses (Table 11-1)
Anemia Resulting From Decreased Production of Erythrocytes or Hemoglobin (Nonregenerative Anemia)
Nutritional Deficiency.
Chronic Disease.
Red Cell Aplasia
Myelophthisic Anemia.
Pathogenesis
Anemic Hypoxia
Autoimmune Hemolytic Anemia
Hemolysis
Methemoglobinemia and Oxidative Damage
Clinical Findings
Clinical Pathology
Hematology
Bone Marrow
Collection of Bone Marrow.
Interpretation of Bone Marrow.
Blood-Gas Analysis, Oximetry and Lactate
Arterial Blood-Gas Analysis.
Venous Blood-Gas Analysis.
Methemoglobinemia.
Lactate.
Serum Biochemistry
Other Evaluations
Necropsy Findings
Treatment
Correction of Anemia
Transfusion.
Transfusion Triggers
Collection of Blood for Transfusion
Hematinics.
Supportive Care
Treatment of Autoimmune Hemolytic Anemia
Further Reading
References
Alloimmune Hemolytic Anemia of the Newborn (Neonatal Isoerythrolysis, Isoimmune Hemolytic Anemia of the Newborn)
Etiology
Epidemiology
Horses and Mules
Pathogenesis
Clinical Findings
Horses and Mules
Pigs
Cattle
Clinical Pathology
Necropsy Findings
Treatment
Foals
Transfusion.
Nutritional Support.
Liver Failure and Kernicterus.
Antibiotics.
Piglets
Control
References
Erythrocytosis
References
Abnormal Red Cell Function
References
Disorders of White Cells
Leukopenia
References
Leukocytosis
Reference
Abnormal White Cell Function
Reference
Leukoproliferative Disease (Leukemia, Lymphoma)
Myeloproliferative Diseases
Further Reading
References
Lymphoproliferative Disease
Plasmacytoma (Multiple Myeloma)
Lymphoma and Lymphosarcoma
Ruminants and Pigs.
Horses
Etiology and Epidemiology.
Clinical Signs.
Clinical Pathology.
Treatment.
Further Reading
References
Bovine Neonatal Pancytopenia (BNP)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
References
Lymphadenopathy (Lymphadenitis)
Reference
Diseases of the Spleen and Thymus
Splenomegaly
Splenic Abscess
Splenic Hematoma, Rupture or Infarction
References
Congenital Anomalies of the Spleen
References
Thymus
References
Immune-Deficiency Disorders (Lowered Resistance to Infection)
Primary Immune Deficiencies
Secondary Immune Deficiencies
References
Amyloidoses
Etiology and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Zoonotic Potential
References
Allergy and Anaphylaxis
Type I
Type II
Type III
Type IV
Treatment
Anaphylaxis and Anaphylactic Shock
Etiology
Pathogenesis
Clinical Findings
Cattle
Sheep, Goats, and Pigs
Horses
Pigs
Clinical Pathology
Necropsy Findings
Treatment
Other Hypersensitivity Reactions
Etiology
Pathogenesis
Clinical Findings
Differential Diagnosis
Treatment
Further Reading
References
Caseous Lymphadenitis of Sheep and Goats
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Sheep
Goats
Source of Infection
Transmission
Risk Factors
Sheep
Age and Sex
Breed
Shearing
Dust
Housing
Dips
Goats
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Culling
Control of Spread
Vaccination
Prevention
Eradication
Further Reading
References
Bovine Farcy
Further Reading
References
Sporadic Lymphangitis (Bigleg, Weed)
Tick-Borne Fever (Anaplasma phagocytophila)
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Experimental Reproduction
Host Risk Factors
Pathogen Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Cattle
Sheep
Goats
Horses
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Anaplasmosis Due to A. Marginale and A. Ovis
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Source and Methods of Transmission
Hematophagous Insect Transmission
Iatrogenic Transmission
Transplacental Transmission
Animal and Environmental Risk Factors
Breed
Nutritional Status
Season
Age at Infection
Geographic Region
Pathogen Risk Factors
Economic Importance
Experimental Reproduction
Pathogenesis
Clinical Findings
Cattle
Sheep and Goats
Clinical Pathology
Hematology
Serology
Molecular Methods
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
General Measures
Movement of Animals
Elimination of Carriers
Outbreaks
Chemotherapy
Vaccination
Killed Vaccines
Live Vaccines
Problems With Live Vaccines
Further Reading
References
Equine Granulocytic Anaplasmosis (Equine Granulocytic Ehrlichiosis, Anaplasma phagocytophilum)
Etiology
Epidemiology
Distribution
Anaplasma and Ixodes Ecology
Animal Risk Factors
Transmission
Morbidity and Mortality
Zoonotic Potential
Pathogenesis
Clinical Signs
Clinical Pathology
Necropsy Examination
Treatment
Control
Further Reading
References
Eperythrozoonosis
Etiology
Epidemiology
Occurrence
Source and Transmission
Sheep
Pigs
Host and Pathogen Risk Factors
Pathogenesis
Clinical Findings
Sheep
Pigs
Cattle
Clinical Pathology
Blood Smears and Hematology
Polymerase Chain Reaction
Serology
Sheep
Pigs
Treatment
Control
Further Reading
References
Bovine Petechial Fever (Ondiri Disease)
Further Reading
Mycoplasma suis Infection in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Pathology
Clinical Signs
Pathology
Diagnosis
Treatment
Control
Further Reading
References
Epizootic Hemorrhagic Disease (Blacktongue)
Etiology
Epidemiology
Occurrence
Host Occurrence
Method of Transmission
Culicoides Species
Other Vectors
Other Methods of Transmission
Host Risk Factors
Wild Ruminants
Cattle
Small Ruminants
Morbidity and Case Fatality
Wild Ruminants
Cattle
Economic Importance
Pathogenesis
Clinical Findings
Deer
Cattle
Clinical Pathology
Virus Isolation
Detection of Antigen or Nucleic Acid
Serologic Tests
Necropsy Findings
Deer
Cattle
Samples for Confirmation of Diagnosis
Treatment
Control
Reduction of Infection Through Vector Abatement
Vaccination
Further Reading
References
Bovine Immunodeficiency-Like Virus
Etiology
Epidemiology
Prevalence of Infection
Methods of Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Detection of Virus.
Serologic Tests.
Necropsy Findings
Further Reading
Enzootic Bovine Leukosis (Bovine Lymphosarcoma)
Etiology
Epidemiology
Prevalence of Infection
Occurrence of Clinical Disease
Methods of Transmission
Direct Contact
Semen, Artificial Insemination, and Embryo Technology
Iatrogenic Transmission
Rectal Palpation
Insects
Congenital Infection
Interspecies Transmission
Source of Infection
Risk Factors
Animal Risk Factors
Genetic Resistance and Susceptibility.
Susceptibility to Other Diseases.
Immune Mechanisms
Environmental and Management Risk Factors
Lack of Biosecurity
Calf Management
Pathogen Risk Factors
Economic Importance
Trade Restrictions
Zoonotic Implications
Other Species
Pathogenesis
Virus and Lesion
Lesions and Clinical Disease
Clinical Findings
Enlargement of the Superficial Lymph Nodes
Digestive Tract Lesions
Cardiac Lesions
Nervous System Involvement
Less Common Lesions
Other Species
Clinical Pathology
Diagnosis of the Presence of Infection With BLV
Serologic Tests
Enzyme-Linked Immunosorbent Assay in Serum or Milk.
Agar Gel Immunodiffusion Test.
Radioimmunoprecipitation Assay.
Radioimmunoassay.
Detection of Virus
Polymerase Chain Reaction.
Differentiation Between Enzootic and Sporadic Bovine Leukosis.
Diagnosis of Persistent Lymphocytosis
Diagnosis of Lymphosarcoma
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
History of Compulsory Eradication Programs in Europe
Eradication Programs
Limitation of Spread of Infection
Prevention of Infection in Calves and Young Stock
Biosecurity
Vaccine
References
Equine Infectious Anemia (Swamp Fever)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Methods of Transmission
Insect Vectors
Other Means of Transmission
Economic Importance
Pathogenesis
Viral Multiplication
Immune Reaction
Anemia and Thrombocytopenia
Persistence of Infection
Summary of Pathogenesis
Clinical Findings
Clinical Pathology
Diagnostic Confirmation
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Ticks That Transmit Protozoan Diseases
Babesiosis (Texas Fever, Redwater Fever, Cattle Tick Fever)
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Bovine Babesiosis
Sheep and Goats
Porcine Babesiosis
Equine Piroplasmosis
Wildlife Babesiosis
Origin of Infection and Transmission
Life Cycle and Development of Babesia
Immunity and Susceptibility to Infection
Innate Immune Mechanisms
Acquired Immune Mechanisms
Immunity.
Risk Factors
Host Factors
Age Resistance.
Environmental Factors
Pathogen Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Acute Cases
Immunology
Clinical Findings
Cattle
Babesia Bovis.
Babesia Bigemina.
Sheep
Wildlife
Other Species
Clinical Pathology
Hematology
Demonstration of the Presence of Babesia
Direct Examination of Blood Smears
Transmission Test
Culture of Babesia
Methods of Detection and Identification of Babesia spp.
Serology
Cattle
Complement Fixation Test.
Immunofluorescence Antibody Test.
ELISA.
Sheep
Necropsy Findings
Treatment
Cattle
Imidocarb (Imizol)
Sheep
Supportive Treatment
Control (Bovine Babesiosis)
Prevention and Biosecurity
Control
Limitation of Prevalence
Vaccination
Live Vaccines
Origin and Purification of Strains.
Attenuation of Parasites
Babesia Bovis.
Babesia Bigemina.
Vaccine Specifications
Frozen Vaccine.
Chilled Vaccine.
Use of Live Vaccine
Cattle Born in Tick-Infested Regions.
Susceptible Cattle Imported Into Vector-Infested Country or Region.
Control of Outbreaks.
Hazards and Precautions of Live Vaccine Use
Severe Reactions.
Lack of Protection.
Vaccination With Subunit Vaccines
Dead Vaccines
Vector Control
Natural Endemic Stability
Control of Babesiosis in Species Other Than Cattle
Further Reading
References
Equine Piroplasmosis
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Impact
Life Cycle and Transmission
Movement of Horses
Pathogenesis
Immunology
Clinical Findings
Clinical Pathology
Microscopic Examination
Serologic and DNA-Based Methods
Necropsy Findings
Treatment
Chemotherapy
Supportive Treatment
Control
References
Nutritional Deficiencies
Iron Deficiency
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Dietary Supplementation
Sows
Veal Calves
Heifer Calf Herd Replacements
Oral Dosing
Intramuscular Injection of Iron Preparations
References
Cobalt Deficiency
Etiology
Epidemiology
Occurrence
Risk Factors
Dietary and Environmental Factors
Ovine White Liver Disease
Hepatic Lipidosis in Goats
Experimental Reproduction of Cobalt Deficiency in Sheep
Pathogenesis
Clinical Findings
Clinical Pathology
Biochemical Criteria to Determine Cobalt and Vitamin B12 Status
Serum and Liver Cobalt and Vitamin B12 Concentrations
Concurrent Serum MMA and Vitamin B12 Concentrations.
Liver Cobalt.
Serum Methylmalonic Acid
Formiminoglutamic Acid
Hematology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Cobalt and Vitamin B12
Cobalt Toxicity
Control
Supplementation of Diet With Cobalt
Top Dressing of Pastures With Cobalt
Cobalt Pellet
Cobalt in Anthelmintics
Vitamin B12 Injections
Further Reading
References
Vitamin B12 Deficiency (Hypocyanocobalaminosis)
References
Vitamin K Deficiency
References
Folic Acid Deficiency (Hypofolicosis)
Reference
Toxins Affecting the Hemolymphatic System
Secondary Copper Poisoning (“Toxemic Jaundice” Complex)
Phytogenous Chronic Copper Poisoning
Hepatogenous Chronic Copper Poisoning
Poisoning by Heliotropium europaeum
Further Reading
Anticoagulant (Dicoumarol, Sweet Clover Poisoning) Plant Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Plant Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Primary
Supportive Treatment
Control
Further Reading
References
Anticoagulant Rodenticide Toxicosis
Etiology
Epidemiology
Occurrence
Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Primary Treatment
Further Treatment
Control
Further Reading
References
Cyanogenic Glycoside Poisoning (Cyanide, Hydrocyanic Acid, Prussic Acid)
Etiology
Toxic Variability
Differences Between Plant Species
Plant Products
Storage
Glycosides
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Plant Risk Factors
Cyanogenic Risk Factors
Environmental Risk Factors
Farm Risk Factors
Pathogenesis
Peracute or Acute Intoxication
Chronic Poisoning
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Field Tests
Samples for Confirmation of Diagnosis
Treatment
Investigational Treatments
Control
Further Reading
References
Nitrate and Nitrite Toxicosis
Etiology
Cattle
Sheep
Swine
Horses
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Species Differences
Dietary Differences
Differences in Susceptibility
Environmental Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Toxic Levels
Ration Dilution
Further Reading
References
Disulfide Plant Toxicosis (Allium spp.[onions] and Brassica spp.)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Farm or Premise Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Red Maple Leaf (Acer rubrum) Toxicosis
References
Neoplasia
Sporadic Bovine Leukosis (Atypical Bovine Leukosis)
Etiology
Epidemiology
Occurrence
Pathogenesis
Clinical Findings
Juvenile or Calf Lymphosarcoma
Thymic Lymphosarcoma
Cutaneous Lymphoma
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
References
Congenital Inherited Diseases
Inherited Bleeding Disorders
Hemophilia
Bovine Factor XI Deficiency
Prekallikrein Deficiency
Inherited Thrombopathia
Glanzmann’s Thrombasthenia in Horses
Afibrinogenemia (Related Diseases Are Hypofibrinogenemia and Dysfibrinogenemia)
References
Familial Polycythemia
Reference
Hemochromatosis
Inherited Anemias
Inherited Dyserythropoiesis and Dyskeratosis (Bovine Congenital Anemia, Dyskeratosis, and Progressive Alopecia)
Inherited Glucose-6-Phosphate Dehydrogenase Enzyme Deficiency
Reference
Porphyrias
References
Inherited Congenital Porphyria
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Inherited Erythrocytic Photoporphyria
References
Inherited Immunodeficiency
Bovine Leukocyte Adhesion Deficiency
Chediak–Higashi Syndrome
References
Inherited Deficiency of Lymphocyte Maturation (Lethal Trait A46, Parakeratosis, Adema Disease)
Fell Pony and Dale Pony Foal Immunodeficiency Syndrome
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Confirmation of Diagnosis
Treatment
Control
References
Inherited Deficiency of Immunoglobulin Synthesis
Inherited Combined Immunodeficiency in Foals of Arabian Breeding
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Differential Diagnosis
Treatment
Control
References
Diseases of Unknown Etiology
Postparturient Hemoglobinuria in Cattle
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Multisystemic, Eosinophilic, Epitheliotropic Disease of Horses
Further Reading
References
12 Diseases of the Respiratory System
Principles of Respiratory Insufficiency
Definitions
Hypoxia
Hypoxic (or Hypoxemic) Hypoxia
Anemic Hypoxia
Circulatory Hypoxia
Histotoxic Anoxia
Consequences of Hypoxia
Compensatory Mechanisms
Carbon Dioxide Retention (Hypercapnia)
Respiratory Failure
Principal Manifestations of Respiratory Insufficiency
Abnormalities in Rate, Depth, and Ease of Breathing
Diseases Causing Dyspnea at Rest or Lack of Exercise Tolerance
Respiratory Tract Disease
Cardiovascular Disease
Diseases of the Blood
Nervous System Diseases
Musculoskeletal Diseases
General Systemic States
Environmental Causes
Miscellaneous Poisons
Abnormal Posture
Normal and Abnormal Breath Sounds
Generation of Breath Sounds
Rebreathing (“Bagging”) Examination
Interpretation of Breath Sounds
Normal Breath Sounds
Abnormal Breath Sounds
Respiratory Noises
Coughing
Cyanosis
Nasal Discharge
Origin
Examination
Nasal Discharge and Location of Lesion
Sampling of Nasal Discharge
Epistaxis and Hemoptysis
Thoracic Pain
Special Examination of the Respiratory System
Auscultation and Percussion
Endoscopic Examination of the Airways (Rhinolaryngoscopy, Tracheobronchoscopy)
Horses
Cattle
Endoscopy of Paranasal Sinuses
Pleuroscopy
Radiography
Magnetic Resonance Imaging
Computed Tomography
Scintigraphy (Nuclear Imaging)
Ultrasonography
Laboratory Evaluation of Respiratory Secretions
Sampling Respiratory Secretions
Nasal Swab
Nasopharyngeal Swabs
Nasal Lavage
Paranasal Sinus Fluid
Guttural Pouch Fluid
Tracheobronchial Secretions
Comparison of Tracheal Aspirates and Bronchoalveolar Lavage Fluid
Tracheal Aspirates
Percutaneous Transtracheal Aspiration
Endoscopic Sampling of Tracheal Secretions
Assessment of Results
Bronchoalveolar Lavage
Endoscopic Bronchoalveolar Lavage
Blind Bronchoalveolar Lavage
Laboratory Assessment of Tracheobronchial Secretions
Diagnostic Value
Thoracocentesis (Pleurocentesis)
Pulmonary Function Tests
Arterial Blood Gas Analysis
Normal Values
Collection of Arterial Blood Gas Samples
Venous Blood Gas Analysis
Pulse Oximetry
Blood Lactate Concentration
Collection and Analysis of Exhaled Breath Condensate
Lung Biopsy
Respiratory Sound Spectrum Analysis
Exercise Testing
Principles of Treatment and Control of Respiratory Tract Disease
Treatment of Respiratory Disease
Respiratory Gas Transport
Oxygen Therapy
Respiratory Stimulants
Mechanical Ventilation
Anti-Inflammatory Therapy
Immunomodulators
Antimicrobial Therapy
Bronchodilator Drugs
Mucolytics, Mucokinetic, and Antitussive Drugs
Surfactant
Surgery
General Nursing Care
Control of Respiratory Disease
Importance of Diagnosis
Management Techniques
Housing Facilities
Vaccines
Environmental Control
Further Reading
References
Diseases of the Upper Respiratory Tract
Rhinitis
Etiology
Cattle
Horses
Sheep and Goats
Pigs
Pathogenesis
Clinical Findings
“Summer Snuffles”
Familial Allergic Rhinitis
Mycotic Rhinitis
Endoscopic Examination
Clinical Pathology
Necropsy Findings
Treatment
References
Nasal Discharge
Neoplasms
Enzootic Nasal Adenocarcinoma
Progressive Ethmoidal Hematomas in Equids
Clinical Findings
Treatment
References
Epistaxis and Hemoptysis
Etiology
Clinical Examination
Treatment
Further Reading
References
Pharyngitis
References
Laryngitis, Tracheitis, Bronchitis
Etiology
Cattle
Sheep
Horses
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Traumatic Laryngotracheitis, Tracheal Compression, and Tracheal Collapse
References
Diseases of the Lung Parenchyma
Pulmonary Congestion and Edema
Etiology
Primary Pulmonary Congestion
Secondary Pulmonary Congestion
Pulmonary Edema
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Special Diseases
References
Pulmonary Hypertension
Causes
References
Atelectasis
References
Pulmonary Hemorrhage
Cattle
References
Pulmonary Emphysema
Etiology
Cattle
Horses
All Species
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Pneumonia
Etiology
Cattle
Pigs
Horses
Sheep
Goats
All Species
Epidemiology
Pathogenesis
Pulmonary Defense Mechanisms
Respiratory Mucociliary Clearance
Large Particles in Upper Respiratory Tract
Cough Reflex
Small Particles Into Lower Respiratory Tract
Species Susceptibility
Development of Pneumonia
Clinical Findings
Medical Imaging
Clinical Pathology
Respiratory Secretions
Thoracocentesis
Hematology
Serology
Fecal Samples
Necropsy
Necropsy Findings
Treatment
Antimicrobial Therapy
Mass Medication
Other Drugs
Supportive Therapy and Housing
References
Acute Respiratory Distress Syndrome
Further Reading
References
Aspiration Pneumonia
References
Lipid Pneumonia
Esophageal Obstruction
References
Meconium Aspiration Syndrome
Reference
Dusty Feed
Drowning
Reference
Pulmonary Abscess
Etiology
Primary Diseases
Secondary Diseases
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Diseases of the Pleural Cavity and Diaphragm
Hydrothorax and Hemothorax
Etiology
Hydrothorax
Hemothorax
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Pleuritis (Pleurisy)
Etiology
Pigs
Cattle
Sheep and Goats
Horses
Other Causes
Pathogenesis
Clinical Findings
Pleural Pain
Pleuritic Friction Sounds
Subcutaneous Edema
Pleural Effusion
Recovery
Medical Imaging
Ultrasonography
Pleuroscopy
Prognosis
Clinical Pathology
Thoracocentesis (Pleurocentesis)
Hematology
Necropsy Findings
Treatment
Antimicrobial Therapy
Drainage and Lavage of Pleural Cavity
Fibrinolytic Therapy
References
Pneumothorax
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Diaphragmatic Hernia
Clinical Findings
References
Diseases of the Bovine Respiratory Tract
Enzootic Nasal Granuloma of Cattle (Bovine Atopic Rhinitis)
References
Tracheal Stenosis of Feedlot Cattle
Caudal Vena Caval Thrombosis (Posterior Vena Caval Thrombosis) and Embolic Pneumonia in Cattle
References
Cranial Vena Caval Thrombosis
Reference
Acute Undifferentiated Bovine Respiratory Disease
Definition of the Problem
Etiology
Role of Etiologic Agents
Clinical Case Definition and Epidemiology
Clinical Case Definition
What Is the Clinical Disease That Is Present in the Affected Animals?
Which Animals Are Affected?
Where Are the Affected Animals?
When Were the Animals Affected?
Occurrence
Risk Factors
Diagnosis and Differential Diagnosis
Individual Animal Diagnosis
Herd-Level Diagnosis
Clinical Pathology
Antemortem Diagnostic Procedures
Nasal/Nasopharyngeal Swabs
Transtracheal Wash/ Bronchoalveolar Lavage
Serology
Serum Biochemistry and Hematology
Other Procedures
Postmortem Samples
Bacterial Culture and Antimicrobial Sensitivity
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Interpretation of Results of Clinical Pathology and Necropsy Findings
Treatment
Control
Mass Medication or Metaphylaxic Antimicrobial Use
Management of Risk Factors
Vaccines
Selection of Vaccines
Efficacy of Vaccines
Pasteurella Vaccines
Histophilus somni (Formerly Haemophilus somnus) Vaccine
Viral Vaccines
Further Reading
References
Pneumonic Pasteurellosis of Cattle (Shipping Fever Pneumonia)
Etiology
Epidemiology
Occurrence
Morbidity and Mortality
Economic Importance
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Pathogen Risk Factors
Immune Mechanisms
Method of Transmission
Pathogenesis
Colonization of Upper and Lower Respiratory Tract
Virulence Factors and Cellular and Humoral Reactions
Experimental Pneumonic Pasteurellosis
Synergism Between Pathogens
Clinical Findings
Feedlot
Early Identification of Affected Animals in Feedlots
Close Physical Examination
Clinical Pathology
Bacterial Culture
Serum Biochemistry and Hematology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Choice of Antimicrobial
Antimicrobial Sensitivity
Medication of Feed and Water Supplies
Antiinflammatory Agents
Failure to Respond
Control
Management Strategies
Preconditioning Programs
Weaning Procedures
Creep Feeding
Conditioning Programs
Feeding Newly Arrived Cattle
Vaccines
General Comments
Pasteurella Vaccines
Leukotoxin Extract Vaccine
Passive Immunity to Mannheimia haemolytica
Evaluation of Efficacy of Mannheimia haemolytica Vaccines
Adverse Vaccine Reactions
Histophilus somni Vaccines
Viral Vaccines
Antimicrobial Metaphylaxis
Mass Medication of Feed and Water Supplies
Further Reading
References
Diseases of the Respiratory Tract Associated With Mycoplasma spp.
Sheep
Goats
Cattle
Horses
References
Contagious Bovine Pleuropneumonia (Lung Sickness, CBPP)
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Source of Infection
Methods of Transmission
Risk Factors
Animal Risk Factors
Immune Mechanisms
Management Risk Factors
Pathogen Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Acute Form
Chronic and Subacute Forms
Clinical Pathology
Culture and Nucleic Acid Recognition
Immunologic Tests
Serologic Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Removal of Sources of Infection
Clinical Surveillance
Pathologic Surveillance
Serologic Surveillance
Vaccination
Antimicrobial Use
Disease Control on an Area Basis
World Organization for Animal Health (OIE)—CBPP Status for Countries or Regions
Further Reading
References
Mycoplasma bovis Pneumonia, Polyarthritis, Mastitis, and Related Diseases of Cattle
Etiology
Epidemiology
Occurrence
Economic Importance
Risk Factors
Pathogen Risk Factors
Animal Risk Factors
Environmental Risk Factors
Methods of Transmission
Pathogenesis
Clinical Findings
Chronic Pneumonia and Polyarthritis Syndrome
Arthritis
Otitis Media/Interna
Clinical Pathology
Culture
DNA Probe and Polymerase Chain Reaction
Immunohistochemistry
Enzyme-Linked Immunosorbent Assay
Serology
Sample Collection and Handling
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Biosecurity
Metaphylactic Use of Antimicrobials
Vaccines
Further Reading
References
Enzootic Pneumonia of Calves
Etiology
Bovine Respiratory Syncytial Virus
Bovine Coronavirus
Parainfluenza-3 Paramyxovirus
Mycoplasma bovis and Mycoplasma spp.
Mixed Viral and Other Pathogen Infections
Bacteria
Epidemiology
Occurrence
Dairy Calves
Beef Calves
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Pathogen Risk Factors
Bovine Respiratory Syncytial Virus
Bovine Coronavirus
Parainfluenza-3 Virus
Mixed Flora
Economic Importance
Pathogenesis
Viruses
Parainfluenza-3
Mycoplasma
Bacteria
Clinical Findings
Clinical Pathology
Isolation of Pathogens
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Adjunctive Therapy
Correction of Adverse Environmental Conditions
Control
Environmental and Management Practices
Ideal Environmental Conditions
Calf Barns or Hutches
Vaccines and Immunization
Further Reading
References
Bovine Respiratory Syncytial Virus
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Occurrence of Clinical Disease
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogen Risk Factors
Immune Mechanisms
Pathogenesis
Experimental Reproduction of BRSV Pneumonia
Clinical Findings
Clinical Pathology
Virus Isolation or Detection
Serology
Arterial Blood Gas Analysis and Blood L-Lactate Concentration
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Corticosteroids and Nonsteroidal Antiinflammatory Agents
Control
Vaccines and Immunization
Further Reading
References
Infectious Bovine Rhinotracheitis (Red Nose), Bovine Herpesvirus-1 Infection
Etiology
Epidemiology
Prevalence of Infection and Occurrence of Disease
Wildlife
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Age and Breed Susceptibility
Environmental and Management Risk Factors
Pathogen Risk Factors
Immune Mechanisms
Colostral Immunity
Economic Importance
Pathogenesis
Respiratory Disease
Encephalitis
Abortion
Latency
Predisposition to Pneumonia
Reproductive Failure
Bovine Mastitis
Clinical Findings
Rhinitis (Red Nose), Tracheitis, and Conjunctivitis
Ocular Form of IBR
Systemic Disease in Newborn Calves
Abortion
Clinical Pathology
Virus Isolation or Detection
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Control
Natural Exposure or Vaccination
Natural Exposure
Biosecurity
Closed Herd
Vaccination
Rationale for Vaccination
Temperature-Sensitive BHV-1 Modified Live Vaccine.
Disadvantages of Modified Live Vaccines.
Shedding of Virus by Vaccinated Animals.
Inactivated Vaccines.
Subunit Vaccines.
Combination or Multivalent Vaccines.
Immunization and Latency.
Vaccination Programs in Herds
Beef Breeding Herds.
Feedlot Cattle.
Dairy Cattle.
Eradication
Eradication Using Marker Vaccines.
Loss of Certification.
Further Reading
References
Lungworm in Cattle
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Atypical Interstitial Pneumonia of Cattle (Acute Bovine Respiratory Distress Syndrome, Acute Pulmonary Emphysema, and Edema)
Etiology
Ingestion of Excessive Amounts of d,l-Tryptophan With the Forage
Hypersensitivity to Molds
Inhalation of Toxic Gases and Fumes
Parasitic Infestation
Mycotoxicosis and Plant Poisonings
Melengestrol Acetate
Bacterial and Mycoplasma spp. Infections
Viral Infections
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Grazing Management
Inhibition of 3-Methylindole Production in Rumen
Other Forms of AIP
Further Reading
References
Diseases of the Ovine and Caprine Respiratory Tract
Enzootic Nasal Adenocarcinoma of Sheep and Goats (Enzootic Nasal Tumor)
References
Contagious Caprine Pleuropneumonia
Etiology
Epidemiology
Occurrence
Transmission
Agent
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Chronic Enzootic Pneumonia of Sheep (Chronic Nonprogressive Atypical Pneumonia, Summer Pneumonia, Proliferative Exudative Pneumonia)
Etiology
M. ovipneumoniae
B. parapertussis
Parainfluenza-3 (PI-3) Virus
Bovine Respiratory Syncytial Virus
Other Agents
Epidemiology
Occurrence
Environmental Risk Factors
Economic Importance
Clinical Findings
Necropsy Findings
Treatment and Control
Further Reading
References
Ovine Progressive Pneumonia (Maedi, Maedi-Visna)
Etiology
Epidemiology
Occurrence
Host Range
Prevalence
Transmission
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Detection of Antigen
Serologic Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Test and Cull
Segregated Rearing
Flock Biosecurity and Other Control Methods
Vaccination and Genetic Selection
Further Reading
References
Ovine Pulmonary Adenocarcinoma (Jaagsiekte, Pulmonary Adenomatosis)
Etiology
Epidemiology
Occurrence
Animal and Environmental Risk Factors
Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Nasal Bots Infestation
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Diagnosis
Treatment
Control
Recommendation
References
Lungworm Infestation in Sheep and Goats
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
References
Diseases of the Equine Respiratory Tract
Abnormalities of the Upper Respiratory Tract of Horses
Further Reading
References
Palatal Dysfunction (Instability, Dorsal Displacement of the Soft Palate)
Palatal Instability and Intermittent Dorsal Displacement of the Soft Palate During Exercise
Clinical Signs
Treatment
Persistent Dorsal Displacement of the Soft Palate
Clinical Signs
Treatment
References
Diseases of the Guttural Pouches (Auditory Tube Diverticulum, Eustachian Tube Diverticulum)
Guttural Pouch Empyema
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
References
Guttural Pouch Mycosis
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Guttural Pouch Tympany
Etiology and Epidemiology
Clinical Findings
Treatment
References
Other Guttural Pouch Diseases
Reference
Diseases of the Epiglottis and Arytenoids
Aryepiglottic Fold Entrapment (Epiglottic Entrapment)
Clinical Signs
Treatment
Epiglottic Retroversion During Exercise
Epiglottitis
Subepiglottic Cysts
Arytenoid Chondritis
Treatment
Mucosal Lesions of the Arytenoid Cartilages
Axial Deviation of the Aryepiglottic Folds
Retropharyngeal Lymphadenopathy
Clinical Signs
Treatment
References
Recurrent Laryngeal Neuropathy (Roarers)
Etiology
Epidemiology
Prevalence
Pathogenesis
Clinical Findings
Necropsy Findings
Diagnostic Confirmation
Treatment
References
Equine Pleuropneumonia (Pleuritis, Pleurisy)
Etiology
Epidemiology
Risk Factors
Pathogenesis
Clinical Signs of Acute Disease
Clinical Signs in Chronic Disease
Complications
Prognosis
Clinical Pathology
Diagnostic Confirmation
Necropsy Findings
Treatment
Antimicrobial Treatment
Thoracic Drainage
Pleural Lavage
Fibrinolytic Therapy
Supportive Therapy
Control
References
Acute Broncho-Interstitial Pneumonia in Foals
Necropsy Findings
Treatment
Control
References
Chronic Interstitial Pneumonia in Foals
Further Reading
Reference
Interstitial Pneumonia in Adult Horses
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Exercise-Induced Pulmonary Hemorrhage of Horses (Bleeders)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Exercise-Induced Pulmonary Hemorrhage and Performance
Physical Examination
Tracheobronchoscopy
Radiography
Prognosis
Clinical Pathology
Examination of Airway Secretions or Lavage Fluid
Necropsy
Treatment
Prevention of Stress Failure of the Pulmonary Capillaries
Reducing Pulmonary Capillary Pressure
Increasing Alveolar Inspiratory Pressure
Interstitial Inflammation and Bronchial Angiogenesis
Excessive Bleeding
Coagulopathy and Fibrinolysis
Platelet Function
Capillary Integrity
Summary of Treatment Options
Prevention and Control
Further Reading
References
Recurrent Airway Obstruction (Heaves)
Etiology
Genetics
Environmental Factors
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Season
Housing and Hay Feeding
Pathogenesis
Inflammatory and Immune Responses
Mucus
Airway Function and Gas Exchange
Clinical Findings
Clinical Pathology and Special Examinations
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Diagnostic Confirmation
Treatment
Antiinflammatory Drugs
Bronchodilator Drugs
Other Drugs
Integrated Therapy
Control
Further Reading
References
Pasture-Associated Heaves (Pasture-Associated Recurrent Airway Obstruction of Horses)
Further Reading
References
Synchronous Diaphragmatic Flutter in Horses (Thumps)
Reference
Rhodococcus equi Pneumonia of Foals
Etiology
Epidemiology
Occurrence
Transmission
Zoonotic Implications
Pathogenesis
Clinical Findings
R. equi pneumonia
Extrapulmonary manifestations of R. equi infection
Prognosis
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Streptococcus zooepidemicus Infection
Further Reading
References
Strangles
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Animal Risk Factors
Importance
Pathogenesis
Clinical Findings
Acute Disease
Complications
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Diagnostic Confirmation
Treatment
Control
Prevention of Transmission
Enhanced Resistance
Further Reading
References
Glanders
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Source of Infection and Transmission
Experimental Reproduction
Host and Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Acute Disease
Chronic Disease
Pulmonary Form of Disease
Nasal Form of Disease
Skin Form of Disease (“Farcy”)
Clinical Pathology
Mallein Test
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Viral Infections of the Respiratory Tract of Horses
Parainfluenza-3 Virus
Equine Adenovirus Infection
Reovirus
References
Equine Influenza
Etiology
Viral Evolution
Persistence in the Environment
Epidemiology
Occurrence
Origin of Infection and Transmission
Risk Factors
Animal Factors
Management Factors
Meteorologic Factors
Economic Importance
Zoonotic Potential
Australian Outbreak (2007)
Pathogenesis
Clinical Findings
Clinical Pathology
Serology
Rapid Detection of Virus
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Immunity and Vaccination
Vaccines
Objective
Timing
Foals
Racehorses and Show Horses
Schedule
Control Measures
Further Reading
References
Equine Rhinitis (Equine Rhinitis Virus)
References
Equine Viral Rhinopneumonitis (Equine Herpesvirus-1 and -4 Infections)
Etiology
Epidemiology
Occurrence
Method of Transmission
Risk Factors
Immunity
Age
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Control
Vaccination
Hygiene
References
Equine Multinodular Pulmonary Fibrosis
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Necropsy Findings
Specimen for Laboratory Diagnosis
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Equine Hendra Virus Infection
Etiology
Epidemiology
Transmission
Zoonotic Potential
Clinical Signs
Clinical Pathology
Necropsy
Treatment and Control
References
Pulmonary and Systemic Aspergillosis (Aspergillus spp.)
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
References
Rhinosporidosis
References
Lungworm in Horses
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Reference
Diseases of the Swine Respiratory Tract
Progressive Atrophic Rhinitis (Conchal Atrophy of Swine)
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Method of Transmission
Risk Factors
Animal Risk Factors
Immune Mechanisms
Pathogen Factors
Environmental Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Culture and Detection of Bacteria
Serology
Antigen Detection
Radiography
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Eradication
Reduction of Infection
Mass Medication
Vaccination
Further Reading
References
Facial Necrosis (Facial Pyemia)
Further Reading
Bordetella Rhinitis
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Immunity
Treatment
Control
References
Pleuropneumonia of Pigs Associated With Actinobacillus pleuropneumoniae
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Pathogen Factors
Biotypes
Serotypes
Virulence Factors
Apx Toxins
Animal Risk Factors
Immune Mechanisms
Environmental and Management Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Culture of Organism
Serotype of Organism
Detection of Antigen
Serology
Necropsy Findings
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Antimicrobial Sensitivities
Antimicrobials in Experimental Disease
Mass Medication of Feed
Control
Control by Management
Eradication
Vaccination
References
Mycoplasma Pneumonia (Mycoplasma hyopneumoniae)
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Immune Mechanisms
Pathogen Risk Factors
Environmental and Management Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Serologic Tests
Detection of Organism
Herd Certification
Necropsy Findings
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Control
Control by Eradication
Specific Pathogen-Free or Minimal-Disease Pigs
Isolated Farrowing
Minimal Disease Herds
Reinfection of Herds
Antimicrobial Prophylaxis
Low-Level Disease
Medication of Breeding Stock
Source of Feeder Pigs
Vaccination
Further Reading
References
Porcine Respiratory Disease Complex and Mycoplasmal Pneumonia of Pigs
Etiology
Epidemiology
Combination and Interaction of Environmental Risk Factors
Economic Losses and Importance
Clinical Signs
Pathology
Treatment
Monitoring
Control
All-in, All-Out
Buildings
Production
Sick Pigs
Diagnosis
Active Control
References
Porcine Cytomegalic Virus (Inclusion-Body Rhinitis, Generalized Cytomegalic Inclusion-Body Disease of Swine)
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
Differential Diagnosis
Treatment
Control
Reference
Swine Influenza
Introduction
Etiology
H1N1—Classical
H1N1—Others
H1N1 Avian-Like
H1N1 Humanlike Viruses in Pigs
H1N1-Pandemic 2009
H1N2
H3N2—Classical
H3N2—Novel
Other Viruses
Epidemiology
Occurrence
Seasonality
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogen Factors
Methods of Transmission
Immunity
Zoonotic Implications
Pathogenesis
Clinical Findings
Concurrent Infections
Clinical Pathology
Experimental Infections
Serologic Tests
Detection of Virus
Oral Fluids
Antigen Detection
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Vaccination
Inactivated Vaccines
Modified Live
Newer Options
Further Reading
References
Porcine Respiratory Coronavirus
Etiolgy
Epidemiology
Pathogenesis
Clinical Signs
Lesions
Diagnosis
Treatment
Control
References
Lungworm in Pigs
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Respiratory System Toxicoses
Furan (Ipomeanol and 3-Methylindole) Toxicosis
Further Reading
References
Galegine Toxicoses
Reference
Manure Gas Poisoning and Confinement Effects
Etiology
Pathogenesis
Clinical Findings
Necropsy Findings
Control
Further Reading
References
Plants Causing Pulmonary Disease (Unidentified Toxins)
Neoplastic Diseases of the Respiratory Tract
Pulmonary and Pleural Neoplasms
Clinical Findings
Treatment
Further Reading
References
Congenital and Inherited Diseases of the Respiratory Tract
Congenital Defects
References
Index
How to Use This Book
For Example
Guidelines for Selection and Submission of Necropsy Specimens for Confirmation of Diagnosis
Volume 2
Front Cover
Veterinary Medicine
Volume 2_Copyright page
Volume 2_Table of contents
13 Diseases of the Urinary System
Introduction
Principles of Renal Insufficiency
Renal Insufficiency and Renal Failure
Causes of Renal Insufficiency and Uremia
Pathogenesis of Renal Insufficiency and Renal Failure
Clinical Features of Urinary Tract Disease
Abnormal Constituents of the Urine
Variations in Daily Urine Flow
Polyuria
Oliguria and Anuria
Pollakiuria
Abdominal Pain and Painful and Difficult Urination (Dysuria and Stranguria)
Morphologic Abnormalities of Kidneys and Ureters
Palpable Abnormalities of the Bladder and Urethra
Acute and Chronic Renal Failure
Uremia
Special Examination of the Urinary System
Tests of Renal Function and Detection of Renal Injury
Collection of Urine Samples
Tests of Urine Samples
Specific Gravity
pH
Net Acid Excretion
Hematuria
Hemoglobinuria
Myoglobinuria
Ketonuria
Glucosuria
Proteinuria
Casts
Cells and Pyuria
Bacteriuria
Crystalluria
Enzymuria
Tests of Serum
Serum Urea Nitrogen and Creatinine Concentration
Glomerular Filtration Rate
Tests of Urine and Serum
Urine Osmolality to Serum Osmolality Ratio
Water Deprivation Test
Renal Clearance Studies
Fractional Clearance
Summary of Renal Function Tests
Diagnostic Examination Techniques
Ultrasonography
Endoscopy
Renal Biopsy
Test of Uroperitoneum and Bladder Rupture
Radiography
Cystometry and Urethral Pressure Profile
Computed Tomography
Principles of Treatment of Urinary Tract Disease
Fluid and Electrolytes
Antimicrobial Agents
Further Reading
References
Diseases of the Kidney
Glomerulonephritis
Reference
Pyelonephritis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Reference
Nephrosis
Ischemic Nephrosis
Etiology
Acute Renal Ischemia
Chronic Renal Ischemia
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Toxic Nephrosis
Etiology
Toxins
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Renal Tubular Acidosis
Nephrogenic Diabetes Insipidus
Distal Renal Tubular Acidosis (Type I)
Proximal Renal Tubular Acidosis (Type II)
Further Reading
References
Hemolytic Uremic–Like Syndrome
Hydronephrosis
Interstitial Nephritis
Embolic Nephritis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Infectious Diseases of the Kidney
Leptospirosis
Etiology
Epidemiology
Risk Factors
Animal Risk Factors
Serovars and Species Susceptibility
Pathogen Risk Factors
Environmental and Management Risk Factors
Occurrence and Prevalence of Infection
Cattle
Farmed Deer
Sheep and Goats
Pigs
Horses
Economic Importance
Zoonotic Implications
Methods of Transmission
Leptospiruria
Wildlife as Source of Infection
Portal of Entry of Organism
Pathogenesis
Acute Form
Septicemia, Capillary Damage, Hemolysis, and Interstitial Nephritis
Abortion
Encephalitis
Subacute and Occult Forms
Periodic Ophthalmia (Recurrent Uveitis) in the Horse
Pulmonary Hemorrhage
Immune Mechanisms
Clinical Findings
Cattle
Acute Leptospirosis Associated With pomona
Subacute Leptospirosis Associated With L. pomona
Chronic Leptospirosis Associated With L. pomona
Leptospirosis Associated With L. hardjo
Pigs
Sheep and Goats
Horses
Abortion
Periodic Ophthalmia
Neonatal Foal Disease
Clinical Pathology
General Considerations
Serologic and Related Tests
Aqueous Humor Antibody
Demonstration or Culture of Organism
Culture of Urine
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Blood Transfusions
Control
Biosecurity and Biocontainment
Eradication
Hygiene
Vaccination
Cattle
Bovine Leptospiral Vaccines and Their Efficacy
Swine
Vaccination and Antimicrobial Strategies
Further Reading
References
Bovine Pyelonephritis
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Environmental Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Urinary Disease in Swine
Glomerulonephritis
Nephrosis
Embolic Nephritis
Interstitial Nephritis
Other Conditions
Porcine Cystitis and Pyelonephritis
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for the Confirmation of Diagnosis
Diagnosis
Treatment
Control
Further Reading
References
Kidney Worm Disease in Pigs Caused by Stephanurus dentatus
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Toxic Agents Affecting the Kidney
Citrinin Toxicosis
Further Reading
References
Ethylene Glycol Toxicosis
Further Reading
References
Ochratoxins (Ochratoxicosis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Factors
Human Risk Factors
Farm or Premise Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnosis
Treatment
Control
Further Reading
References
Plant Poisonings Caused by Known Toxins
Iforrestine
Plant Poisonings From Unidentified Toxins
Uremia, Nephrosis—With High Blood Urea Nitrogen
Polydipsia, Polyuria
Red Urine Caused by A Pigmented Substance From the Plant
Cystitis
Fungi Lacking Identified Toxins
Further Reading
Renal Neoplasia
References
Congenital and Inherited Renal Diseases
Renal Hypoplasia
References
Polycystic Kidneys
Reference
Renal Dysplasia
References
Renal Lipofucinosis of Cattle
References
Equine Renal Cortical Tubular Ectasia
Reference
Diseases of the Ureters, Bladder, and Urethra
Ectopic Ureter and Ureteral Defects
Paralysis of the Bladder and Overflow Incontinence
Reference
Eversion of the Bladder
Reference
Patent Urachus
Rupture of the Bladder (Uroperitoneum)
References
Uroperitoneum in Foals
Etiology
Epidemiology
Pathophysiology
Clinical Signs
Imaging
Clinical Pathology
Necropsy Findings
Treatment
Prevention and Control
Reference
Cystitis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Urolithiasis in Ruminants
Etiology
Epidemiology
Species Affected
Nidus Formation
Precipitation of Solutes
Factors Favoring Concretion
Miscellaneous Factors in the Development of Urolithiasis
Composition of Calculi
Risk Factors for Obstructive Urolithiasis
Occurrence
Pathogenesis
Clinical Findings
Obstruction of the Urethra by a Calculus
Rupture of Urethra or Bladder
Clinical Pathology
Urinalysis
Serum Biochemistry
Abdominocentesis and Needle Aspirate of Subcutaneous Tissue
Ultrasonography
Radiography
Necropsy Findings
Treatment
Prevention
Further Reading
References
Urolithiasis in Horses
Further Reading
Reference
Urethral Tears in Stallions and Geldings
Urethral Defects
Further Reading
Reference
Urinary Bladder Neoplasms
References
Bovine Enzootic Hematuria
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Primary
Supportive
Control
Further Reading
References
Diseases of the Prepuce and Vulvovaginal Area
Enzootic Posthitis (Pizzle Rot, Sheath Rot, Balanoposthitis) and Vulvovaginitis (Scabby Ulcer)
Etiology
Epidemiology
Host Occurrence
Sheep
Cattle
Source of Infection and Transmission
Host and Environmental Risk Factors
Experimental Reproduction
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy and Diagnostic Confirmation
Treatment
Control
Further Reading
14 Diseases of the Nervous System
Introduction
Posture and Gait
Sensory Perceptivity
Mental State
Principles of Nervous Dysfunction
Modes of Nervous Dysfunction
Excitation (Irritation) Signs
Release of Inhibition Signs
Paresis or Paralysis Caused by Tissue Damage
Nervous Shock
Clinical Manifestations of Diseases of the Nervous System
Altered Mentation
Excitation States
Mania
Frenzy
Aggressive Behavior
Depressive States
Depression Leading to Coma
Syncope
Narcolepsy (Catalepsy)
Compulsive Walking or Head Pressing
Aimless Wandering
Involuntary Movements
Tremor
Tics
Tetany
Convulsions
Involuntary Spastic Paresis
Abnormal Posture and Gait
Posture
Vestibular Disease
Gait
Ataxia
Cerebellar Disease
Spinal Cord Disease
Paresis and Paralysis
Neurogenic Muscular Atrophy
Altered Sensation
Blindness
Central or Peripheral Blindness
Abnormalities of the Autonomic Nervous System
Special Examination of the Nervous System
Neurologic Examination
Signalment and Epidemiology
History
Head
Behavior
Mental Status
Head Position and Coordination
Cranial Nerves
Olfactory Nerve (Cranial Nerve I)
Optic Nerve (Cranial Nerve II)
Oculomotor Nerve (Cranial Nerve III)
Trochlear Nerve (Cranial Nerve IV)
Trigeminal Nerve (Cranial Nerve V)
Abducent Nerve (Cranial Nerve VI)
Facial Nerve (Cranial Nerve VII)
Vestibulocochlear Nerve (Cranial Nerve VIII)
Glossopharyngeal Nerve (Cranial Nerve IX) and Vagus Nerve (Cranial Nerve X)
Spinal Accessory Nerve (Cranial Nerve Xi)
Hypoglossal Nerve (Cranial Nerve XII)
Posture and Gait
Neck and Forelimbs
Spinal Reflexes of the Thoracic Limbs
Trunk and Hindlimbs
Tail and Anus
Palpation of the Bony Encasement of the Central Nervous System
Collection and Examination of Cerebrospinal Fluid
Collection of Cerebrospinal Fluid
Collection From the Lumbosacral Cistern
Collection From the Atlantooccipital Cistern (Cisterna Magna)
Cerebrospinal Fluid Pressure
Analysis of Cerebrospinal Fluid
Examination of the Nervous System With Serum Biochemical Analysis
Arterial Plasma Ammonia Concentration
Further Reading
References
Examination of the Nervous System With Imaging Techniques
Radiography
Computed Tomography
Magnetic Resonance Imaging
Further Reading
References
Ultrasonography
Endoscopy (Rhinolaryngoscopy)
Ophthalmoscopy
Electromyography
Electroencephalography
Electroretinography
Brainstem Auditory Evoked Potentials
Intracranial Pressure Measurement
Kinetic Gait Analysis
Further Reading
References
Diffuse or Multifocal Diseases of the Brain and Spinal Cord
Responses of Central Nervous System to Injury
Cerebral Hypoxia
Etiology
Cerebral Hypoxia Secondary to General Hypoxia
Cerebral Hypoxia Secondary to Intracranial Lesion
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Treatment
Increased Intracranial Pressure, Cerebral Edema, and Brain Swelling
Etiology
Vasogenic Edema
Cytotoxic Edema
Interstitial Edema
Pathogenesis
Cerebral Edema and Brain Swelling
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Hydrocephalus
Etiology
Congenital Hydrocephalus
Acquired Hydrocephalus
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Meningitis
Etiology
Cattle
Sheep
Horses
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Cerebrospinal Fluid
Hematology
Necropsy Findings
Treatment
Further Reading
References
Encephalitis
Etiology
All Species
Cattle
Sheep
Goats
New World Camelids
Horses
Pigs
Pathogenesis
Clinical Findings
Clinical Pathology
Hemogram
Serology
Cerebrospinal Fluid
Necropsy Findings
Treatment
Further Reading
References
Epilepsy
Treatment
Further Reading
Reference
Myelitis
Encephalomalacia
Etiology
All Species
Ruminants
Horses
Ruminants and Horses
Neurotoxic Mycotoxins
Pigs
Pathogenesis
Metabolic and Circulatory
Intoxications and Toxic-Infectious Diseases
Nutritional Diseases
Hereditary, Familial, and Idiopathic Degenerative Diseases
Polioencephalomalacia and Leukoencephalomalacia
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Myelomalacia
References
Focal Diseases of the Brain and Spinal Cord
Traumatic Injury to the Brain
Etiology
Pathogenesis
Concussion
Contusion
Laceration
Experimental Traumatic Craniocerebral Missile Injury
Clinical Findings
Diagnosis
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Brain Abscess
Etiology
Hematogenous Spread
Local Spread
Pathogenesis
Clinical Findings
Clinical Pathology
Cerebrospinal Fluid
Hematology
Imaging
Electroencephalography
Necropsy Findings
Treatment
Further Reading
References
Tumors of the Central Nervous System
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Central Nervous System–Associated Tumors
Further Reading
Reference
Metastatic Tumors of the Central Nervous System
Central Nervous System–Associated Masses
References
Plant Toxins Affecting the Nervous System
Cannabinoids
Cynanchoside
Diterpenoid (Kaurene) Glycosides (Atractyloside, Carboxyatractyloside, Parquin, Carboxyparquin, and Wedeloside)
Stypandrol
Tropane Alkaloids
Tutin
Further Reading
References
Indole Alkaloids
β-Carboline Indoleamine Alkaloid Poisoning
Further Reading
References
Indolizidine Alkaloid Toxicosis (Locoism, Peastruck)
Castanospermine Poisoning
Swainsonine Poisoning
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Neurogenic Quinolizidine Alkaloids (Lupinus Spp.)
Etiology
Epidemiology
Clinical Findings
Pathology
Further Reading
Nitrocompound Plant Toxicosis (Milk Vetch)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Acute Poisoning
Chronic Poisoning
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Piperidine Alkaloid Plant Toxicosis
Etiology
Conium
Epidemiology
Pathogenesis
Clinical Findings
Cynapine
Nicotiana
Lobeline
Further Reading
References
Corynetoxins (Tunicaminyluracils) (Annual Ryegrass Staggers, Flood Plain Staggers, Stewart Range Syndrome)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Plant Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Miscellaneous Plant Toxins Affecting the Nervous System (Unidentified Toxins)
Further Reading
References
Fungal Toxins Affecting the Nervous System
Further Reading
Reference
Tremorgenic Mycotoxins
Aspergillus-Associated Mycotoxins
Bermudagrass Staggers
Claviceps-Associated Mycotoxins (Paspalum or Dallis Grass Staggers)
Neotyphodium-Associated Mycotoxins (Perennial Ryegrass Staggers)
Penicillium-Associated Mycotoxins
Further Reading
References
Miscellaneous Fungal Toxins Affecting the Nervous System (Unidentified Toxins)
Black Soil Blindness
Nervous Signs
Further Reading
Other Toxins Affecting the Nervous System
Inorganic Toxins Affecting the Nervous System
Lead Toxicosis (Plumbism)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Cattle
Buffalo
Sheep
Horses
Environmental Risk Factors
Farm or Premise Risk Factors
Human and Public Health Risk Factors
Transmission (Sources of Lead)
Pathogenesis
Clinical Findings
Cattle
Sheep
Horses
Pigs
Clinical Pathology
Hematology
Blood Lead
Milk Lead
Fecal Lead
Urinary Lead
δ-ALA-D
Erythrocyte Protoporphyrin
Plasma δ-Aminolevulinic Acid
Necropsy Findings
Liver and Kidney Lead
Cattle
Horses
Samples for Confirmation of Diagnosis
Treatment
Calcium Versenate
Succimer (Dimercaptosuccinic Acid)
Thiamine Hydrochloride
Magnesium Sulfate
Rumenotomy
Control
Further Reading
References
Mercury Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Boron Toxicosis
Further Reading
Bromide Toxicosis
Further Reading
References
Organic Toxins Affecting the Nervous System
Anthelmintic Toxicosis
Commonly Used Anthelmintics
Older Anthelmintics
Currently Used Anthelmintics
Amino-Acetonitrile Derivatives (Monepantel)
Benzimidazoles (Albendazole, Fenbendazole, and Thiabendazole) and Probenzimidazoles (Febantel, Netobimin, etc.)
Albendazole, Cambendazole, and Parbendazole
Fenbendazole
Thiabendazole
Cyclic Octadepsipeptides (Emodepside)
Imidazothiazoles (Levamisole)
Macrocyclic Lactones (Ivermectin, Moxidectin, and Doramectin)
Miscellaneous (Piperazine and Clorsulon)
Piperazine
Clorsulon
Praziquantel/Epsiprantel
Salicylanilides/Substituted Phenols (Closantel, Rafoxanide, and Oxyclozanide)
Tetrahydropyrimidines (Pyrantel and Morantel)
Older Anthelmintics
Carbon Tetrachloride
Hexachloroethane
Hexachlorophene
Nicotine
Phenothiazine
Sumicidin
Tetrachlorethylene
Further Reading
References
Macrocyclic Lactone (Ivermectin, Moxidectin, etc.) Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogenesis
Clinical Findings
Necropsy Findings
Treatment
Control
Further Reading
References
Organophosphorus Compounds and Carbamate Insecticides
Etiology
Epidemiology
Occurrence
Source of Toxin
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Transmission
Pathogenesis
Cholinesterase Inactivation
Organophosphorus-Induced Delayed Neurotoxicity
Clinical Findings
Acute Poisoning
Cattle, Sheep, and Goats
Acute Toxicosis
Delayed Neurotoxicity
Pigs
Acute Toxicosis
Delayed Neurotoxicity
Horses
Acute Toxicosis
Delayed Neurotoxicity Syndrome
Miscellaneous Signs of Organophosphorus Poisoning
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Industrial Organophosphates
Rotenone Toxicosis
Further Reading
References
Organochlorine Insecticides
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Farm or Premise Risk Factors
Environmental Risk Factors
Human Risk Factors
Transmission
Method of Application
Formulation Used
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Further Reading
Sodium Fluoroacetate (Compound 1080) Toxicosis
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology/ Necropsy Findings
Treatment/Control
Further Reading
References
Molluscicide Toxicosis
Metaldehyde
Methiocarb
Further Reading
References
Strychnine
Further Reading
Diseases of the Cerebrum
Psychoses, Neuroses, and Stereotypy
Crib-Biting and Windsucking
Weaving
Box Walking
Farrowing Hysteria in Sows
Tail-Biting, Ear-Chewing, and Snout-Rubbing in Pigs
Head-Shaking in Horses
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Tail-Biting in Swine
Etiology
Epidemiology
Risk Factors
Factors Increasing Biting
Clinical Findings
Clinical Pathology
Necropsy
Treatment
Control
Further Reading
References
Bacterial Diseases Primarily Affecting the Cerebrum
Enterotoxemia Associated With Clostridium Perfringens Type D (Pulpy Kidney, Overeating Disease)
Etiology
Epidemiology
Occurrence
Experimental Reproduction
Animal and Management Risk Factors
Sheep
Calves
Goats
Horses
Pathogenesis
Clinical Findings
Lambs
Adult Sheep
Calves
Goats
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Reduction in Food Intake
Antitoxin
Vaccination
Further Reading
References
Focal Symmetric Encephalomalacia
Etiology
Epidemiology
Clinical Findings
Necropsy Findings
Treatment and Control
Further Reading
References
Cerebrospinal Angiopathy
Further Reading
Viral Diseases Primarily Affecting the Cerebrum
Rabies
Etiology
Epidemiology
Occurrence
Europe
United States
Canada
Africa
South America, Latin America, and the Caribbean
Distribution of Virus Variants
Methods of Transmission
Animal Vectors
Seasonal Spread
Latent Infection
Zoonotic Implications
Economic Importance
Pathogenesis
Clinical Findings
Cattle
Sheep and Goats
Horses
Pigs
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Prevention of Exposure to the Virus
Preexposure Vaccination of Humans
Postexposure Vaccination of Humans
Postexposure Vaccination of Domestic Animals
Vaccination of Domestic Animals
Vaccines
Vaccination of Wildlife
Quarantine and Biosecurity
Further Reading
References
Pseudorabies (Aujeszky’s Disease)
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogen Factors
Methods of Transmission
Within Herds
Between Herds
Within an Area
Latency
Other Species
Immune Mechanisms
Economic Importance
Pathogenesis
Clinical Findings
Pigs
Cattle, Sheep, and Goats
Clinical Pathology
Serology
Serum Neutralization Test
Enzyme-Linked Immunosorbent Assay
Detection of Virus
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Strategies Available
Economics of Control and Eradication
Determination of Prevalence of Infection
Depopulation and Repopulation
Test and Removal
Offspring Segregation
Control Programs in Effect
Vaccines and Vaccination
Vaccines
Pregnant Sows
Growing and Finishing Pigs
Marker or Subunit Vaccines
References
Sporadic Bovine Encephalomyelitis (Buss Disease and Transmissible Serositis)
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Morbidity and Case–Fatality Rates
Method of Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Hematology
Detection of Agent
Serology
Necropsy Findings
Treatment
Control
References
Border Disease (Hairy Shaker Disease of Lambs, Hairy Shakers, Hypomyelinogenesis Congenita)
Etiology
Epidemiology
Occurrence
Source of Infection
Transmission
Host Risk Factors
Experimental Reproduction
Economic Importance
Pathogenesis
Nonpregnant Sheep
Pregnant Sheep
Infection in Early Pregnancy
In Midpregnancy
Infection in Late Pregnancy
Goats
Enteric Disease
Clinical Findings
Conformation
Fleece
Neurologic Dysfunction
Growth Rate
Reproductive Performance
Clinical Pathology
Detection of Persistently Infected Sheep
Abortion
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Visna
Etiology
Epidemiology
Occurrence
Experimental Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
Caprine Arthritis Encephalitis
Etiology
Epidemiology
Geographic Occurrence
Host Risk Factors
Breeds
Age
Method of Transmission
Colostrum and Milk
Other Perinatal Transmission
Contact Transmission
Other Routes
Experimental Reproduction
Economic Importance
Pathogenesis
Clinical Findings
Joints
Brain
Udder
Clinical Pathology
Serologic Testing
Other Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Prevention of Perinatal Transmission
Test and Segregate/Cull
Vaccination and Genetic Selection
Further Reading
References
Ovine Encephalomyelitis (Louping-Ill)
Etiology
Epidemiology
Occurrence
Geographic Occurrence
Host Occurrence
Transmission
Tick Transmission
Nontick Transmission
Host and Environmental Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
West Nile, Kunjin, and Murray Valley Encephalitis
Etiology
Epidemiology
Distribution
Viral Ecology
Transmission
Animal Risk Factors
Morbidity and Case Fatality
Zoonotic Implications
Pathogenesis
Clinical Findings
Other Species
Clinical Pathology
Serologic Tests
Identification of West Nile Virus
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Japanese Encephalitis
Etiology
Epidemiology
Clinical Signs
Clinical Pathology
Necropsy Findings
Zoonosis
Samples for Confirmation of Diagnosis
Differential Diagnosis
Treatment and Control
References
Eastern and Western Equine Encephalomyelitis
Etiology
Epidemiology
Distribution
Viral Ecology
Western Equine Encephalomyelitis
Eastern Equine Encephalomyelitis
Animal Risk Factors
Morbidity and Case Fatality
Zoonotic Implications
Pathogenesis
Clinical Findings
Pigs
Ratites and Pheasants
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Vaccination
Protection From Insects
Zoonotic Aspects of Control
Further Reading
References
Venezuelan Equine Encephalomyelitis
Etiology
Epidemiology
Distribution
Viral Ecology
Animal Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Vaccination
Protection From Insects
References
Equid Herpesvirus-1 Myeloencephalopathy, Abortion, and Neonatal Septicemia
Etiology
Epidemiology
Occurrence
Method of Transmission
Cycling of Infection
Risk Factors
Immunity
Economic Importance
Pathogenesis
Clinical Findings
Myeloencephalopathy
Abortion
Neonatal Viremia and Septicemia
Respiratory Disease
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Prevention of Infection
Vaccination
Subdivision of Horses on a Farm
Minimize Risk of Introduction of Infection
Prevention of Reactivation of Latent Infection
Control of Outbreaks
Control of Outbreaks of Myeloencephalopathy
Abortion
Rapid Diagnosis
Prevention of Spread
Further Reading
References
Peruvian Horse Sickness Virus
References
Powassan Virus
Reference
Nigerian Equine Encephalitis
Main Drain Virus Encephalitis
Reference
Borna Disease
Further Reading
References
Teschovirus Infections
Serotypes
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Pathogenesis
Clinical Findings
Acute Viral Encephalomyelitis (Teschen Disease)
Subacute Viral Encephalomyelitis (Talfan Disease)
Clinical Pathology
Serology
Detection of Virus
Necropsy Findings
Samples for Confirmation of Diagnosis
Immunity
Treatment
Control
Further Reading
References
Prion Diseases Primarily Affecting the Cerebrum
Introduction
Bovine Spongiform Encephalopathy (Mad Cow Disease)
Etiology
Epidemiology
Occurrence
Geographic Occurrence
Occurrence in Cattle
Great Britain
Herd Type
Northern Ireland and Republic of Ireland
European Continent and Iberian Peninsula
North America
Japan
Age Incidence
Other Species
Method of Natural Transmission
Ingestion of Meat-and-Bone Meal
Born-After-the-Ban
Non–Feed-Borne Transmission
Vertical Transmission
Risk for Occurrence of Disease in Countries
Experimental Reproduction
Infectivity of Tissues
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Signs and Passive Surveillance
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Detection of BSE in Surveillance and Control Programs
Control of BSE in Cattle
Measures to Protect Human Health
Further Reading
References
Bovine Spongiform Encephalopathy and Sheep
Experimental Transmission
Pathogenesis
Clinical Signs
Disposition of Disease-Associated PrP
Diagnosis
Strain Tying
Control
References
Scrapie
Etiology
Epidemiology
Occurrence
Geographic Occurrence and Incidence
Host Occurrence
Age
Breed
Methods of Transmission
Sources and Routes of Infection
Horizontal Transmission
Vertical Transmission
Environment
Iatrogenic Transmission
Genetics
Risk Factors
Exposure Factors
Age at Exposure
Infection Status of Parents
Goats
Experimental Reproduction
Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Incubation
Early Signs
Advanced Cases
Clinical Pathology
Necropsy Findings
Control
Individual Flocks
National Eradication
Flock Eradication
Genetic Control and National Programs
Further Reading
References
Chronic Wasting Disease
Further Reading
Parasitic Disease Primarily Affecting the Cerebrum
Coenurosis (Gid, Sturdy)
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
References
Halicephalobus
References
Metabolic Diseases Primarily Affecting the Cerebrum
Polioencephalomalacia (Cerebrocortical Necrosis) of Ruminants
Etiology
Thiamine Inadequacy
Excess Dietary Sulfur
Epidemiology
Occurrence
Morbidity and Case Fatality
Risk Factors
Dietary Risk Factors
Thiamine Inadequacy
Thiaminases
Sulfur-Induced Polioencephalomalacia
Other Dietary Circumstances
Deprivation of Feed and Water.
Phalaris Aquatica “PEM-Like” Sudden Death in Sheep and Cattle.
Pathogenesis
Thiamine Inadequacy Polioencephalomalacia
Sulfate-Induced Polioencephalomalacia
Acute Cerebral Edema and Laminar Necrosis
Clinical Findings
Cattle
Sheep
Clinical Pathology
Thiamine Inadequacy Polioencephalomalacia.
Brain Imaging Function.
Sulfate-Induced Polioencephalomalacia
Ruminal Hydrogen Sulfide Measurement.
Brain Function.
Necropsy Findings
Treatment
Thiamine Hydrochloride
Outbreak Management
Sulfur-Induced Polioencephalomalacia
Control
Thiamine Supplementation
Feeding Roughage
Sulfate Toxicity PEM
Further Reading
References
Thiamine Deficiency (Hypothiaminosis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Bracken Fern (P. aquilinum) and Horsetail (E. arvense) Poisoning in the Horse
Experimental Syndromes
Clinical Pathology
Necropsy Findings
Treatment
Control
Thiaminase Toxicosis
Etiology
Epidemiology
Occurrence
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Salt Toxicity (Sodium Chloride Toxicosis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Farm Risk Factors
Environmental Risk Factors
Pathogenesis
Acute Poisoning
Chronic Poisoning
Clinical Findings
Subclinical Salt Poisoning
Acute Salt Poisoning
Subacute Poisoning
Chronic Salt Poisoning
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Acute Toxicity
Chronic Toxicity
Control
Further Reading
References
Vitamin A Deficiency (Hypovitaminosis A)
Etiology
Epidemiology
Primary Vitamin A Deficiency
Ruminants on Pasture
Maternal Deficiency
Adequacy of Supplements
Feedlot Cattle
Pigs
Horses
Secondary Vitamin A Deficiency
Pathogenesis
Night Vision and Ocular Abnormalities
Cerebrospinal Fluid Pressure
Bone Growth
Epithelial Tissues
Embryologic Development
Immune Mechanisms
Clinical Findings
Night Blindness
Xerophthalmia
Ocular Abnormalities
Changes in the Skin
Body Weight
Reproductive Efficiency
Nervous System
Paralysis
Convulsions
Blindness
Congenital Defects
Other Diseases
Anasarca.
Clinical Pathology
Plasma Vitamin A
Plasma Retinol
Plasma Carotene
Hepatic Vitamin A
Cerebrospinal Fluid
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Vitamin A
Hypervitaminosis A
Control
Dietary Requirement
Supplementation Method
Parenteral Injection
Oral Vitamin A
References
Nicotinic Acid Deficiency (Hyponiacinosis)
References
Pyridoxine (Vitamin B6) Deficiency (Hypopyridoxinosis)
Reference
Pantothenic Acid Deficiency (Hypopantothenosis)
Reference
Metabolic and Toxic Encephalomyelopathies
Abnormalities of Consciousness and Behavior
Abnormality Characterized by Tremor and Ataxia
Convulsions
Ataxia Apparently Caused by Proprioceptive Defect
Involuntary Spastic Contraction of Large Muscle Masses
Tremor, Incoordination, and Convulsions
Paresis or Paralysis
Further Reading
References
Inherited Diseases Primarily Affecting the Cerebrum
Inherited Congenital Hyrdocephalus
Cattle
Sheep
Horses
Pigs
References
Inherited Hydranencephaly and Arthrogryposis
Inherited Prosencephaly
Inherited Multifocal Symmetric Encephalopathy
Maple Syrup Urine Disease (Branched-Chain Keto Acid Dehydrogenase Deficiency)
Reference
Inherited Citrullinemia
Inherited Neonatal Spasticity
Doddler Calves
Inherited Idiopathic Epilepsy of Cattle
Familial Narcolepsy
Further Reading
Reference
Congenital and Inherited Encephalomyelopathies
Inherited Lysosomal Storage Diseases
Mannosidosis
α-Mannosidosis
β-Mannosidosis
References
Gangliosidosis
GM1 Gangliosidosis
GM2 Gangliosidosis
References
Gaucher Disease Type 2
Reference
Bovine Mucopolysaccharidosis Type IIIB
Reference
Sphingomyelinase Deficiency (Niemann–Pick Disease Type a) in Cattle
Reference
Globoid Cell Leukodystrophy (Galactocerebrosidosis)
Inherited Nervous System Abiotrophies
Further Reading
Neuronal Ceroid Lipofuscinosis
References
Congenital Necrotizing Encephalopathy in Lambs
Reference
Lavender Foal Syndrome
References
Inherited Hypomyelinogenesis (Congenital Tremor Syndromes of Piglets)
Further Reading
Diseases Primarily Affecting the Cerebellum
Inherited Cerebellar Defects
Cerebellar Hypoplasia
Cerebellar Atrophy of Lambs (Daft Lamb Disease 1)
Star-Gazing Lambs (Daft Lamb Disease 2)
Hereditary Lissencephaly and Cerebellar Hypoplasia in Churra Lambs
References
Inherited Ataxia of Calves
Familial Convulsions and Ataxia in Cattle
Inherited Congenital Spasms of Cattle
Cerebellar Abiotrophy
Cattle
Sheep
Alpaca
Horses
Pigs
References
Diseases Primarily Affecting the Brainstem and Vestibular System
Otitis Media/Interna
Pigs
Calves and Lambs
Horses
Treatment
Further Reading
References
Listeriosis
Etiology
Epidemiology
Occurrence
Geographic
Seasonal
Host
Source of Infection
Silage
Transmission
Risk Factors
Host Management Risk Factors
Pathogen Risk Factors
Experimental Reproduction
Zoonotic Implications
Pathogenesis
Encephalitis/Meningitis
Spinal Myelitis
Mastitis
Enteritis
Clinical Findings
Listerial Encephalitis/Meningitis
Sheep
Cattle
Goats
Listerial Abortion
Cattle
Sheep and Goats
Abortion Caused by Listeria Ivanovii
Septicemic Listeriosis
Mastitis
Spinal Myelitis
Keratoconjunctivitis, Uveitis
Enteritis in Sheep
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Central Nervous System Listeriosis
Septicemia and Abortion
Enteritis
Treatment
Control
Further Reading
References
Diseases Primarily Affecting the Spinal Cord
Traumatic Injury
Etiology
Physical Trauma
Parasitic Invasion
Local Ischemia of the Spinal Cord
Pathogenesis
Clinical Findings
Fracture of the Cervical Vertebrae in Horses
Spondylosis in Bulls
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
Reference
Spinal Cord Compression
Etiology
Tumors
Vertebral Body or Epidural Abscess
Bony Lesions of Vertebra
Vertebral Subluxation or Compressive Myelopathy
Ataxia in Horses
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
References
Back Pain in Horses
Parasitic Diseases Primarily Affecting the Spinal Cord
Equine Protozoal Myeloencephalitis
Etiology
Epidemiology
Risk Factors
Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Cerebrospinal Nematodiasis (Elaphostrongylosis)
References
Setaria
Further Reading
References
Toxic Diseases Primarily Affecting the Spinal Cord
Stringhalt
References
Inherited Diseases Primarily Affecting the Spinal Cord
Spastic Paresis of Cattle (Elso Heel)
Further Reading
References
Inherited Congenital Myoclonus (Hereditary Neuraxial Edema)
Inherited Spinal Dysmyelination
Inherited Neurodegeneration (Shaker Calf Syndrome)
Inherited Spinal Dysraphism
References
Inherited Congenital Posterior Paralysis
Inherited Bovine Degenerative Axonopathy
Degenerative Axonopathy of Tyrolean Grey Cattle
Reference
Central and Peripheral Axonopathy of Maine Anjou (Rouge-Des- Prés) Cattle
Inherited Progressive Degenerative Myeloencephalopathy (Weaver Syndrome) of Brown Swiss Cattle
Inherited Progressive Ataxia
Inherited Spinal Myelinopathy
Inherited Periodic Spasticity of Cattle
Neuraxonal Dystrophy
Neuraxonal Dystrophy of Sheep (Segmented Axonopathy)
Neuraxonal Dystrophy of Horses
References
Caprine Progressive Spasticity
Inherited Spontaneous Lower Motor Neuron Diseases
Reference
Inherited Spinal Muscular Atrophy
Inherited Hypomyelinogenesis (Congenital Tremor of Pigs)
Porcine Congenital Progressive Ataxia and Spastic Paresis
Reference
Equine Degenerative Myeloencephalopathy (Equine Neuraxonal Dystrophy)
References
Equine Cervical Vertebral Compressive Myelopathy (Wobbler, “Wobbles,” Foal Ataxia, Equine Sensory Ataxia, Cervical Vertebral Instability)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogenesis
Clinical Findings
Neurologic Examination
Ancillary Diagnostic Tests
Radiographic Examination
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Equine Motor Neuron Disease
Further Reading
References
Diseases Primarily Affecting the Peripheral Nervous System
Etiology
Inflammatory
Degenerative
Traumatic
Metabolic and Nutritional
Toxic
Tumors
Autonomic Nervous System
Further Reading
References
Tetanus
Etiology
Epidemiology
Occurrence
Case–Fatality Rate
Source of Infection
Transmission
Animal Risk Factors
Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Passive Immunity
Tetanus Antitoxin
Active Immunity
References
Botulism
Etiology
Epidemiology
Occurrence
Source of Infection
Forage Botulism
Carrion-Associated Botulism
Wound Botulism
Toxicoinfectious Botulism
Experimental Reproduction
Risk Factors
Animal Risk Factors
Environment Risk Factors
Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Cattle and Horses
Sheep
Goats
Pigs
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Tick Paralysis
Further Reading
References
Ovine “Kangaroo Gait” and Fenugreek Staggers
Etiology
Epidemiology
Occurrence
Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
Reference
Polyneuritis Equi (Cauda Equina Syndrome)
References
Scandinavian Knuckling Syndrome (Acquired Equine Polyneuropathy)
References
Peripheral Nerve Sheath Tumors
Clinical Findings
Clinical Pathology
Treatment
References
15 Diseases of the Musculoskeletal System
Principal Manifestations of Musculoskeletal Disease
Lameness
Abnormal Posture and Movement
Deformity
Muscle and Tendon Defects
Defects of the Skeleton
Joint Defects
Spontaneous Fractures
Painful Aspects of Lameness
Relief of Musculoskeletal Pain
References
Examination of the Musculoskeletal System
Analysis of Gait and Conformation
Close Physical Examination
Radiography
Ultrasonography
Arthrocentesis and Synovial Fluid Interpretation
Arthroscopy
Serum Biochemistry and Enzymology
Muscle Biopsy
Infrared Thermography
Nuclear Scintigraphy
Magnetic Resonance Imaging
Computed Tomography
Nutritional History
Environment and Housing
References
Diseases of Muscles
Myasthenia (Skeletal Muscle Asthenia)
Myopathy
Etiology and Epidemiology
Enzootic Nutritional Muscular Dystrophy
Exertional or Postexercise Rhabdomyolysis
Equine Atypical Myopathy (Seasonal Pasture Myopathy)
Equine Polysaccharide Storage Myopathy
Metabolic
Degenerative Myopathy
Inherited Myopathies
Toxic Agents
Ischemia
Neurogenic
Neoplasms
Pathogenesis
Primary Myopathy
Myoglobinuria
Muscle Enzymes
Exertional Rhabdomyolysis
Types of Muscle Fiber Affected
Secondary Myopathy Resulting From Ischemia
Neurogenic Atrophy of Muscle
Clinical Findings
Primary Myopathy
Acute Nutritional Myopathy
Tying-Up
Postanesthetic Myositis
Exertional Rhabdomyolysis
Hyperkalemic Periodic Paralysis
Secondary Myopathy Resulting From Ischemia
Neurogenic Atrophy
Dystrophy of the Diaphragmatic Muscles
Diagnosis
Muscle-Derived Serum Enzymes
Muscle Biopsy
Myoglobinuria
Necropsy Findings
Treatment
Control
Further Reading
Reference
Myopathy of Horses
Etiology
Pathogenesis
Diagnosis
Treatment
Control
Further Reading
Reference
Myositis
Acute Myositis of Limb Muscles
Injection-Site Lesions in Cattle
Injection-Site Clostridial Infections in Horses
Reference
Diseases of Bones
Osteodystrophy
Etiology
Nutritional Causes
Calcium, Phosphorus, and Vitamin D
Copper Deficiency
Other Nutritional Causes
Chemical Agents
Inherited and Congenital Causes
Physical and Environmental Causes
Tumors
Pathogenesis
Rickets
Osteomalacia
Osteodystrophia Fibrosa
Osteoporosis
Osteodystrophy of Chronic Fluorosis
Congenital Defects of Bone
Clinical Findings
Diagnosis
Necropsy Findings
Treatment
References
Hypertrophic Osteopathy (Marie’s Disease)
References
Osteomyelitis
Etiology and Pathogenesis
Osseous Sequestration in Cattle
Osteomyelitis Secondary to Trauma
Inflammation of Bone Marrow
Clinical Findings
Diagnosis
Necropsy Findings
Treatment
Further Reading
References
Tail-Tip Necrosis in Beef Cattle
Risk Factors
Treatment
Control
Reference
Laminitis of Horses
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Phenotype
Hyperinsulinemia
Supporting Limb Laminitis
Importance
Pathogenesis
Clinical Findings
Prognosis
Clinical Pathology
Necropsy Findings
Treatment
Treatment of Inciting Process or Disease
Cryotherapy or Digital Cooling
Analgesics and Antiinflammatory Drugs
Vasodilatory Drugs
Anticoagulants
Mechanical Support
Promotion of Healing
Chronic Laminitis
Control
Further Reading
References
Laminitis in Ruminants and Swine
Etiology
Epidemiology
Occurrence
Risk Factors
Cattle and Sheep
Pigs
Importance
Pathogenesis
Clinical Findings
Cattle and Sheep
Pigs
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Diseases of Joints
Degenerative Joint Disease (Osteoarthropathy) and Osteochondrosis
Etiology and Epidemiology
Nutritional Causes
Toxic Causes
Steroid Induced
Biomechanical Trauma
Aging Process
Osteochondrosis
Pathogenesis
Primary Osteoarthropathy
Secondary Osteoarthropathy
Osteochondrosis
Clinical Findings
Diagnosis
Joint Fluid
Radiography
Arthroscopy
Necropsy Findings
Treatment
Nonsteroidal Antiinflammatory Agents and Opioids
Intraarticular Steroids
Chondroprotective Agents
Hyaluronic Acid
Polysulfated Glycosaminoglycan
Other Treatments
Control and Prevention
Further Reading
References
Septic Arthritis Synovitis
Etiology and Epidemiology
Calves
Lambs
Foals
Piglets
Cattle
Sheep and Goats
Horses
Pigs
All Species
Pathogenesis
Calves With Experimentally Induced Infectious Arthritis
Lambs With Streptococcus dysgalactiae Polyarthritis
Foals With Septicemia
Horses
Clinical Findings
Diagnosis
Arthrocentesis
Analysis of Joint Fluid
Culture of Joint Fluid
Serology of Joint Fluid
Radiography
Ultrasonography
Arthroscopy
Nuclear Scintigraphy
Magnetic Resonance Imaging and Computed Tomography
Necropsy Findings
Treatment
Parenteral Antimicrobials
Intraarticular Antimicrobials
Lavage of Joint
Arthroscopy
Surgical Drainage and Arthrotomy
Arthrodesis or Artificial Ankylosis
Physical Therapy
Antiinflammatory Agents and Adjunctive Therapy
Prognosis for Survival and Athletic Use in Horses With Septic Arthritis
Control
Further Reading
References
Lameness in Pigs and Degenerative Joint Disease (Osteochondrosis, Osteoarthrosis, Epiphysiolysis and Apophysiolysis, Leg Weakness in Pigs)
Leg Disorders
Lameness
Age-Related Changes in Pigs
Young Pigs
Growers and Finishers
Older Animals
Adults
Osteochondrosis
Etiology
Epidemiology
Occurrence
Risk Factors
Nutrition and Rate of Growth
Genetic and Breed Predisposition
Type of Flooring
Exercise and Confinement
Economic Importance
Pathogenesis
Radiologic Monitoring of Lesions
Clinical Findings
Epiphysiolysis
Apophysiolysis
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Infectious Diseases of the Musculoskeletal System
Borreliosis (Lyme Borre liosis, Lyme Disease)
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Cattle
Sheep
Horses
Wildlife
Zoonotic Implications
Methods of Transmission
Pathogenesis
Immune Mechanisms
Clinical Findings
Horses
Cattle
Sheep
Clinical Pathology
Detection of Organism
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Malignant Edema, Clostridial Myonecrosis (Gas Gangrene)
Etiology
Epidemiology
Source of Infection
Transmission
Animal and Management Risk Factors
Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
Reference
Blackleg
Etiology
Epidemiology
Occurrence
Source of Infection
Transmission
Risk Factors
Environment Risk Factors
Animal Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Cattle
Sheep
Horses
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Cattle
Sheep
Further Reading
References
Bovine Footrot (Infectious Bovine Pododermatitis, Interdigital Phlegmon, Interdigital Necrobacillosis, Foul in the Foot)
Etiology
Epidemiology
Occurrence
Transmission
Environmental Risk Factors
Host Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Interdigital Dermatitis/Stable Footrot
Verrucose Dermatitis
Traumatic Injury
Treatment
Antimicrobials
Local Treatment
Surgical Drainage
Control
Footbaths
Antibacterials
Vaccination
Further Reading
References
Bovine Digital Dermatitis, Papillomatous Digital Dermatitis of Cattle (Mortellaro’s Disease), Foot Warts, Hairy Foot Warts, “Heel Warts”
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Risk Factors
Host Risk Factors
Immune Mechanisms
Environmental and Management Risk Factors
Pathogen Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Detection of Organism
Serology
Pathology
Treatment
Topical Antimicrobials
Nonantibiotic Topical Formulations
Cleaning the Surface of the Lesion
Bandaging the Lesion
Antimicrobials Parenterally and Topically
Parenteral Antimicrobials
Footbaths
Antibiotics in Footbaths
Treatment Failure
Control
Further Reading
References
Infectious Footrot in Sheep
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Source of Infection
Methods of Transmission
Host Risk Factors
Age and Sex
Breed
Environmental Risk Factors
Climate and Season
Management
Pasture Type
Pathogen Factors
Economic Importance
Pathogenesis
Benign and Virulent Footrot
Clinical Findings
Sheep
Virulent Footrot
Benign Footrot
Scoring Systems
Symptomless Carriers
Goats
Cattle
Clinical Pathology and Necropsy Findings
Treatment and Control
Topical Treatment
Footbathing for Treatment and Control
Zinc Sulfate Solution (10% to 20%)
Formalin Solution (5%)
Copper Sulfate Solution (5%)
Antibiotic Treatment
Vaccination
Multivalent Vaccines
Mono- and Bivalent Vaccines
Summary of Control Procedures in Infected Flocks
Genetic Selection
Eradication
Introduced Sheep
Further Reading
References
Foot Abscess in Sheep
Heel Abscess/Infectious Bulbar Necrosis
Treatment of Heel Abscess
Toe Abscess
Treatment of Toe Abscess
Further Reading
Lamb Arthritis
Chlamydial Polyarthritis
Further Reading
References
Chronic Pectoral and Ventral Midline Abscess in Horses (Pigeon Fever)
External Abscesses
Internal Abscesses
References
Infectious Polyarthritis (Glässer’s Disease, Porcine Polyserositis, and Arthritis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Bacteriology
Histology
Serology
Diagnosis
Treatment
Control
Further Reading
References
Arthritis Resulting From Erysipelas
Erysipelas in Sheep
Arthritis in Lambs
Postdipping Lameness
References
Mycoplasma Hyosynoviae in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Pathology
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
Footrot in Pigs (Bush Foot)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Ross River Virus
Etiology
Epidemiology
Zoonotic Implications
Clinical Signs
Diagnosis
Treatment
Control
References
Nutritional Diseases Affecting the Musculoskeletal System
Selenium and/or Vitamin E Deficiencies
Etiology
Biological Functions of Selenium and Vitamin E (VE)
Selenium
Glutathione Peroxidases and Tissue Peroxidation
Vitamin
Interrelationships Between Selenium and Vitamin E
Epidemiology
Enzootic Nutritional Muscular Dystrophy
Occurrence
Geographic Distribution
Selenium in Soil, Plants, and Animals
Selenium in Soils
Selenium in Plants
Factors Influencing the Availability of Soil Selenium to Plants.
Selenium in Animals
Vitamin E
Polyunsaturated Fatty Acids in Diet
Other Myopathic Agents in Diet
Unaccustomed Exercise
Congenital Nutritional Muscular Dystrophy
Vitamin E–Selenium Deficiency Syndrome
Mulberry Heart Disease
Hepatosis Dietetica
Selenium-Responsive Disorders
Subclinical Selenium Insufficiency
Reproductive Performance
Pigs
Sheep
Cattle
Retained Fetal Placenta
Mammary Gland Function
Transport Stress
Resistance to Infectious Disease
Neutrophil Function
Immune Response
General Resistance
Transfer of Selenium and Vitamin E to the Fetus, Colostrum, and Milk
Selenium
Pigs
Vitamin E
Neonatal Morbidity and Mortality
Mastitis
Blood Abnormalities
Equine Nutritional Myopathy
Equine Degenerative Myeloencephalopathy
Equine Motor Neuron Disease
Generalized Steatitis
Pathogenesis
Nutritional Muscular Dystrophy
Subclinical Selenium Insufficiency
VESD Syndrome and Others
Clinical Findings
Acute Enzootic Muscular Dystrophy
Subacute Enzootic Muscular Dystrophy
Vitamin E/Selenium Deficiency in Pigs
Mulberry Heart Disease
Hepatosis Dietetica
Clinical Pathology
Myopathy
Plasma Creatine Kinase
Aspartate Aminotransferase
Selenium Status
Selenium Status in Horses
Kidney Cortex and Liver
Blood and Milk
Bulk-Tank Milk
Glutathione Peroxidase
Vitamin E Status
Farmed Red Deer
Pigs
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Nutritional Muscular Dystrophy
Control
Provide Selenium and Vitamin E
Maternal Transfer to Newborn
Selenium Is Potentially Toxic
Selenium in Milk Supplies
Dietary Requirement of Selenium
Glutathione Peroxidase Activity
Pigs
Different Methods of Supplementation
Dietary Supplementation
Selenium Dose
Individual Injections
Subcutaneous Injections
Cattle and Sheep.
Pigs.
Farmed Red Deer.
Oral Selenium and Anthelmintics
Pasture Top Dressing
Muscular Dystrophy
Beef Cattle and Sheep
Salt–Mineral Mixture
Dairy Cattle
Selenium
Vitamin E
Selenium-Responsive Reproductive Performance and Growth
Sheep
Weak-Calf Syndrome
Pigs
Horses
Intraruminal Selenium Pellets
Sheep
Cattle
Selenium Toxicity and Residues
Selenium Responsiveness
Further Reading
References
Masseter Myonecrosis
References
Sporadic Exertional Rhabdomyolysis in Horses (Azoturia, Tying-Up)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Dietary Deficiency of Phosphorus, Calcium, and Vitamin D and Imbalance of the Calcium : Phosphorus Ratio
Absorption and Metabolism of Calcium and Phosphorus
References
Calcium Deficiency
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Specific Syndromes
Primary Calcium Deficiency
Secondary Calcium Deficiency
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Phosphorus Deficiency
Etiology
Epidemiology
Geographic Occurrence
Cattle
Sheep and Horses
Pigs
Secondary Phosphorus Deficiency
Pathogenesis
Clinical Findings
Clinical Pathology
Serum Phosphorus
Dietary Phosphorus Content
Bone Ash Concentrations
Necropsy Findings
Treatment
Control
Phosphorus Requirements
Cattle
Dairy Cattle
Environmental Implications of Phosphorus Feeding of Livestock
Beef Cattle
Feedlot Cattle
Pigs
Phosphorus Supplementation
Further Reading
References
Vitamin D Deficiency
Etiology
Epidemiology
Ultraviolet Irradiation
Dietary Vitamin D
Grazing Animals
Animal Risk Factors
Pathogenesis
Maternal Status
Calcium : Phosphorus Ratio
Other Roles for Vitamin D
Clinical Findings
Clinical Pathology
Serum Calcium and Phosphorus
Plasma Vitamin D
Necropsy Findings
Treatment
Control
Supplementation
Injection
Further Reading
References
Vitamin D Intoxication
Samples for Confirmation of Diagnosis
References
Rickets
Etiology
Epidemiology
Calves
Lambs
Pigs
Foals
New World Camelids
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
Osteomalacia
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Ruminants
Pigs
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
References
Osteodystrophia Fibrosa
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Horse
Pigs
Goats
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
“Bowie” or “Bentleg” in Lambs
Degenerative Joint Disease and Osteoarthritis
Further Reading
References
Manganese Deficiency
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
References
Biotin (Vitamin H) Deficiency (Hypobiotinosis)
Cattle
Sheep
Pigs
Biotin Requirements
Pigs
Horses
References
Toxic Agents Affecting the Musculoskeletal System
Hyena Disease of Cattle
Further Reading
Reference
Calcinogenic Glycoside Poisoning (Enzootic Calcinosis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Hypoglycin A Intoxication of Horses (Atypical Myopathy [Myoglobinuria] in Grazing Horses)
References
Plant Poisonings With Known Toxins
Aminopropionitrile
Plant Poisonings With Suspected or Unidentified Toxins
Juglone
Myopathy—With Gait Incoordination, Recumbency, Elevated CPK
Further Reading
Aluminum Toxicosis
Further Reading
References
Fluoride Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Supplementary Feeding of Phosphates
Farm Risk Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Acute Intoxication
Chronic Intoxication
Dental Fluorosis
Osteofluorosis
Other Effects
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Congenital Defects of Muscles, Bones, and Joints
Weakness of Skeletal Muscles
Congenital Hyperplasia of Myofiber
Obvious Absence or Deformity of Specific Parts of the Musculoskeletal System
Fixation of Joints
Cattle
Sheep and Goats
Foals
Piglets
Congenital Arthrogryposis and Hydranencephaly, Akabane Disease, Cache Valley Virus Disease, Schmallenberg Virus
Etiology
Epidemiology
Occurrence
Akabane
Cache Valley
Schmallenberg
Aino and Shamonda
Source of Infection
Akabane
Cache Valley
Schmallenberg
Aino
Host and Environmental Risk Factors
Akabane
Cache Valley
Schmallenberg
Experimental Reproduction
Zoonotic Implications
Pathogenesis
Akabane
Cache Valley
Schmallenberg
Clinical Findings
Akabane
Cache Valley
Schmallenberg
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
Hypermobility of Joints
Inherited Diseases of Muscles
Glycogen Storage Diseases
Generalized Glycogenosis (Glycogen Storage Disease Type II)
Further Reading
Reference
Glycogen Storage Disease Type V (Muscle Glycogen Phosphorylase Deficiency)
References
Inherited Diaphragmatic Muscle Dystrophy
Reference
Congenital Myasthenia Gravis
References
Bovine Familial Degenerative Neuromuscular Disease
Inherited Umbilical Hernia
Myofiber Hyperplasia (Double Muscling, Doppelender, Culard)
Epidemiology
Clinical Findings
Clinical Pathology
Necropsy Findings
References
Inherited Splayed Digits
Inherited Progressive Muscular Dystrophy
Pseudomyotonia of Cattle, Congenital Muscular Dystonia-1
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Congenital Muscular Dystonia Type 1 (CMD1)
Pseudomyotonia (PMT)
Clinical Pathology
Treatment
Necropsy Findings
Control
References
Ovine Humpyback
Further Reading
Myotonia of Goats (Fainting Goats)
Etiology
Epidemiology
Occurrence
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Further Reading
References
Myotonia Congenita and Myotonic Dystrophy
References
Recurrent Exertional Rhabdomyolysis in Thoroughbred and Standardbred Horses
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Diagnostic Confirmation
Treatment AND CONTROL
Control
Further Reading
References
Polysaccharide Storage Myopathy of Horses
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Equine Hyperkalemic Periodic Paralysis
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Necropsy Findings
Treatment
Acute Episodes
Prevention of Episodes
Control
References
Malignant Hyperthermia in Horses
References
Porcine Stress Syndrome (Malignant Hyperthermia)
Etiology
Epidemiology
Prevalence and Occurrence
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Porcine Stress Syndrome (Transport Death)
Malignant Hyperthermia
Pale, Soft, and Exudative Pork
Dark, Firm, and Dry Pork
Back-Muscle Necrosis
Clinical Pathology
Halothane Test
Blood Creatine Kinase Levels
Blood Typing
Pale, Soft, and Exudative Pork
Erythrocyte Osmotic Fragility
Other Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Genetic Selection
Management of Stressors
Pietrain Creeper Pigs
Further Reading
Asymmetric Hindquarter Syndrome of Pigs
Further Reading
Porcine Congenital Splayleg (Splayleg Syndrome in Newborn Pigs)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Treatment
Control
Further Reading
References
Inherited Diseases of Bones
Etiology
Pathogenesis
Clinical Findings
Diagnosis
Treatment and Control
Further Reading
References
Inherited Osteogenesis Imperfecta
Cattle
Sheep
Reference
Inherited Dwarfism
Sheep
Snorter Dwarfs
Inherited Congenital Achondroplasia With Hydrocephalus
Miscellaneous Dwarfs
References
Congenital Osteopetrosis
Reference
Inherited Probatocephaly (Sheepshead)
Inherited Atlanto-Occipital Deformity
Inherited Agnathia
Inherited Displaced Molar Teeth
Inherited Jaw Malapposition
References
Inherited Cranioschisis (Cranium Bifidum)
References
Inherited Craniofacial Deformity
References
Inherited Arachnomelia (Inherited Chondrodysplasia)
Cattle
Sheep
Spider Lamb Syndrome
Inherited Chondrodysplasia in Texel Sheep
References
Complex Vertebral Malformation in Holstein Calves
References
Inherited Reduced Phalanges (Amputates, Acroteriasis, Ectromelia)
Hereditary Hemimelia
Hereditary Peromelia of Mohair Goats
Amputates
References
Inherited Claw Deformity
References
Inherited Multiple Exostosis
Inherited Congenital Hyperostosis (Thick Forelimbs of Pigs)
Inherited Rickets
References
Inherited Taillessness and Tail Deformity
References
Congenital Chondrodystrophy of Unknown Origin (Ccuo, “Acorn” Calves, Congenital Joint Laxity and Dwarfism, Congenital Spinal Stenosis)
References
Inherited Diseases of Joints
Inherited Arthrogryposis (Inherited Multiple Tendon Contracture)
Simple Arthrogryposis
Arthrogryposis With Dental Dysplasia
Arthrogryposis With Palatoschisis
Arthrogryposis With Multiple Defects
Arthrogryposis in Species Other Than Cattle
Inherited Multiple Ankyloses
References
Inherited Patellar Subluxation
Reference
Inherited Hypermobility (Laxity) of Joints
Inherited Hip Dysplasia
Reference
16 Diseases of the Skin, Eye, Conjunctiva, and External Ear
Introduction
Primary/Secondary Lesions
Clinical Signs and Special Examination
Lesions
Abnormal Coloration
Pruritus
Cattle
Sheep
Horses
Pigs
All Species
Secretion Abnormalities of Skin Glands
Abnormalities of Wool and Hair Fibers
Further Reading
Principles of Treatment of Diseases of the Skin
Primary Treatment
Supportive Treatment
Further Reading
Diseases of the Epidermis and Dermis
Pityriasis
Treatment
Hyperkeratosis
References
Parakeratosis
Treatment
Pachyderma
Treatment
Impetigo
Treatment
Urticaria
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Primary Treatment
Supportive Treatment
Further Reading
References
Dermatitis and Dermatosis
Etiology
All Species
Cattle
Sheep and Goats
Horses
Pigs
Special Local Dermatitides
Pathogenesis
Clinical Findings
Pemphigus
Clinical Pathology
Diagnosis
Treatment
Reference
Photosensitization
Etiology and Epidemiology
Type I, Primary Photosensitization
Type II, Photosensitization as a Result of Aberrant Pigment Metabolism
Type III, Hepatogenous Photosensitization
Plants Containing Hepatotoxins
Plants Containing Steroidal Saponins
Congenitally Defective Hepatic Function
Type IV, Photosensitization of Uncertain Etiology
Pathogenesis
Clinical Findings
General Signs
Skin Lesions
Systemic Signs
Nervous Signs
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
Diseases of the Hair, Wool, Follicles, and Skin Glands
Alopecia and Hypotrichosis
Etiology
Failure of Follicles to Develop
Loss of Follicles
Failure of the Follicle to Produce a Fiber
Congenital
Acquired
Loss of Preformed Fibers
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Further Reading
References
Achromotrichia
Leukoderma and Leukotrichia
Etiology
Pathophysiology
Clinical Findings
Clinical Pathology
Treatment
Further Reading
Vitiligo
Etiology
Pathophysiology
Clinical Findings
Clinical Pathology
Treatment
Further Reading
Reference
Seborrhea
Etiology
Clinical Findings
Flexural Seborrhea
Greasy Heel of Cows
Greasy Heel of Horses (Scratches)
Clinical Pathology
Treatment
Folliculitis
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Reference
Diseases of the Subcutis
Subcutaneous Edema (Anasarca)
Etiology
Increased Hydrostatic Pressure
Hypoproteinemic (Hypooncotic) Edema
Increased Blood Vessel Permeability
Fetal Anasarca
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Angioedema (Angioneurotic Edema)
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Subcutaneous Emphysema
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Lymphangitis
Etiology
Cattle
Horse
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Panniculitis
Hematoma
Etiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Primary Treatment
Supportive Treatment
Necrosis and Gangrene
Etiology
Other Causes
Pathogenesis
Clinical Findings
Treatment
Subcutaneous Abscess
Trauma
Hematogenous
Extension
Cutaneous Cysts
Granulomatous Lesions of the Skin
Cattle
Sheep
Horses
Pigs
Reference
Non-Infectious Diseases of the Skin
Insect Bite Hypersensitivity in Horses (Equine Seasonal Allergic Dermatitis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
Reduce Exposure to Biting Midges
References
Seasonal Allergic Dermatitis of Sheep
References
Anhidrosis (Nonsweating Syndrome, Puff Disease, Dry Coat)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Wetness (Maceration)
Reference
Cockle
Control
Further Reading
Wool Slip, Wool Loss
Wool Eating
Further Reading
Idiopathic Nasal/Perioral Hyperkeratotic Dermatosis of Camelids (Mouth Munge)
Clinical Findings
Clinical Pathology
Treatment
References
Bacterial Diseases of the Skin
Methicillin-Resistant Staphylococcus Aureus
Introduction
Human Public Health
Veterinarians
Pig Farmers
Butchers
Animal MRSA
Etiology
Epidemiology
Distribution
Colonization
Transmission
Risk Factors
Pigs
Cattle
Veal Calves
Sheep and Goats
Small Animals
Horses
Other Species
Poultry
Turkeys
Donkeys
Wild Boar
Backyard Pigs
Camels
Fish
Zoo Animals
Other Sources
Holding Areas
State Fairs
Food
Pathogenesis
Pathology
Diagnosis
Control
Further Reading
References
Dermatophilosis (Mycotic Dermatitis Cutaneous Streptotrichosis, Senkobo Disease of Cattle, Lumpy Wool of Sheep)
Etiology
Epidemiology
Occurrence
Geographic Occurrence
Host Occurrence
Source of Infection
Transmission
Environmental and Management Risk Factors
Sheep
Cattle
Temperate Zones
Tropical Zones
Horses
Host Risk Factors
Pathogen Risk Factors
Experimental Disease
Economic Importance
Sheep
Cattle
Zoonotic Implications
Pathogenesis
Clinical Findings
Sheep
Cattle
Horses
Goats
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Sheep
Cattle
Horses
Control
Further Reading
References
Skin Tuberculosis
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Mycobacterium ulcerans Infection (Buruli or Bairnsdale Ulcer)
References
Fleece Rot in Sheep
Etiology
Epidemiology
Occurrence
Environmental and Host Risk Factors
Fleece Characteristics
Experimental Production
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Diagnosis
Control
References
Bolo Disease
Further Reading
Strawberry Footrot of Sheep (Proliferative Dermatitis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
References
Contagious Acne of Horses (Canadian Horsepox, Contagious Pustular Dermatitis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
Exudative Epidermitis (Greasy Pig Disease)
Etiology
Epidemiology
Occurrence
Method of Transmission
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnosis
Treatment
Control
References
Ulcerative Dermatitis (Granulomatous Dermatitis) of Pigs
Reference
Viral Diseases of the Skin
Papillomavirus Infection (Papillomatosis, Warts)
Etiology
Equids
Pigs
Epidemiology
Occurrence
Origin of Infection and Transmission
Animal Risk Factors
Age
Experimental Production
Economic Importance
Pathogenesis
Clinical Findings
Cattle
Goats
Horses
Clinical Pathology
Treatment
Vaccination
Control
Further Reading
References
Sarcoid
Etiology
Epidemiology
Occurrence and Prevalence
Methods of Transmission
Experimental Reproduction
Animal Risk Factors
Environmental Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Further Reading
References
Cowpox and Buffalopox
Etiology
Epidemiology
Occurrence
Origin of Infection and Transmission
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Control
References
Pseudocowpox (Milkers’ Nodule)
Etiology
Epidemiology
Occurrence
Origin of Infection and Transmission
Animal Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Treatment
Control
References
Lumpy Skin Disease (Knopvelsiekte)
Etiology
Epidemiology
Occurrence
Origin of Infection and Transmission
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogen Risk Factors
Experimental Transmission
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Vaccination
Further Reading
References
Sheeppox and Goatpox
Etiology
Epidemiology
Prevalence of Infection
Methods of Transmission
Experimental Reproduction
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Antigen Detection
Serology
Necropsy Findings
Treatment
Control
Further Reading
References
Contagious Ecthyma (Contagious Pustular Dermatitis, Orf, Scabby Mouth, Soremouth)
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Methods of Transmission
Experimental Reproduction
Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Sheep
Goats
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Ulcerative Dermatosis of Sheep
Further Reading
Poxvirus Infections in Horses (Horsepox, Uasin Gishu, Viral Popular Dermatitis, Equine Molluscum Contagiosum)
References
Swinepox
Etiology
Epidemiology
Occurrence
Methods of Transmission
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
Dermatomycoses
Ringworm
Etiology
Epidemiology
Occurrence, Source of Infection, and Transmission
Risk Factors
Pathogen Factors
Environment and Host Factors
Zoonotic Considerations and Economic Importance
Pathogenesis
Clinical Findings
Cattle
Sheep
Horses
Pigs
Clinical Pathology
Treatment
Local Application
Sprays, Washes
Systemic Treatment
Control
Hygiene
Vaccination
Nutrition
Further Reading
References
Mucormycosis
References
Malassezia spp. Dermatitis
References
Sporotrichosis
Etiology
Epidemiology
Pathogenesis, Clinical Findings and Clinical Pathology
Treatment and Control
Reference
Epizootic Lymphangitis (Pseudoglanders, Equine Blastomycosis, Equine Histoplasmosis)
Etiology
Epidemiology
Zoonotic Potential
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Equine Phycomycosis (Swamp Cancer, Pithyosis, Hyphomycosis Destruens, Florida Horse Leech, Bursattee)
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Zoonotic Potential
Pathogenesis and Clinical Findings
Clinical Pathology
Treatment
References
Maduromycosis
Protozoal Diseases of the Skin
Besnoitiosis (Elephant Skin Disease)
Etiology
Epidemiology
Occurrence
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Bovine Besnoitiosis
Acute Stage
Chronic Stage
Caprine Besnoitiosis
Equid Besnoitiosis
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment and Control
Further Reading
References
Nematode Infections of the Skin
Summer Sores in Horses (Habronemosis)
Etiology
Life Cycle
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Rhabditid Dermatitis
Reference
Onchocerciasis (Worm Nodule Disease)
References
Parafilariosis
References
Stephanofilariasis
Cutaneous Myiasis
Blow-Fly Strike of Sheep
Etiology
Life Cycle and Epidemiology
The Fly Population
Susceptibility of Sheep
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Treatment
Control
Reduction of Fly Numbers
Prediction of Risk Periods
Treatment and Prevention
Reducing the Susceptibility of Sheep
Further Reading
References
Screwsworm (Cochliomyia hominivorax and Chrysomyia bezziana)
Etiology
Epidemiology
Life Cycle
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Wohlfahrtiosis (Flesh Fly, Wohlfahrtia magnifica)
Life Cycle and Epidemiology
Pathogenesis
Clinical Pathology and Necropsy Findings
Treatment
Recommendation
References
Mite Infestations
Harvest Mites (Chigger Mites)
Recommendations
References
Itchmites (Psorergates ovis, Psorergates bos)
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Treatment and Control
Demodectic Mange (Follicular Mange)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment and Control
Reference
Sarcoptic Mange (Barn Itch)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment and Control
Recommendation
Reference
Psoroptic Mange (Sheep Scab, Body Mange, Ear Mange)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Sheep
Goats
Horses
Cattle
Clinical Pathology
Treatment and Control
Recommendation
References
Chorioptic Mange (Tail Mange, Leg Mange, Scrotal Mange)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment and Control
References
Ked and Louse Infestations
Sheep Ked (Melophagus ovinus)
Life Cycle
Control
References
Louse Infestations (Pediculosis)
Etiology
Life Cycle and Epidemiology
Sucking Lice
Chewing Lice
Clinical Findings and Diagnosis
Sucking Lice
Chewing Lice
Treatment and Control
Recommendation
References
Miscellaneous Skin Diseases Caused by Flies, Midges, and Mosquitoes
Stable Flies (Stomoxys calcitrans)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Treatment and Control
Recommendation
References
Horse Flies, March Flies or Breeze Flies (Tabanus Spp.), and Deer Flies (Chrysops, Haematopota, and Pangonia Spp.)
Recommendation
Reference
Hypoderma spp. Infestation (Warble Flies)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Macrocyclic Lactone Compounds
Treatment Recommendations
Manual Removal
Control
References
Horn Flies and Buffalo Flies (Haematobia spp.)
Etiology
Life Cycle and Epidemiology
Pathogenesis
Clinical Findings
Treatment and Control
Recommendation
References
Black Flies, Buffalo Gnats (Simuliidae)
Housefly (Muscidae— Musca domestica)
Recommendation
Bush Flies (Musca vetustissima)
Recommendation
Reference
Face Fly (Musca autumnalis)
Recommendation
Head Fly (Hydrotoea irritans)
Reference
Biting Midges (Ceratopogonidae)
References
Mosquitoes (Culicidae)
Tick Infestations
Bacterial, Viral, and Rickettsial Diseases Transmitted by Ticks
Ticks That Cause Direct Losses
Treatment and Control of Tick Infestations
Acaricidal Agents
Organophosphates
Pyrethroids
Macrocyclic Lactones
Use of Resistant Cattle
Pasture Management
Vaccination
Eradication
Further Reading
References
Deficiencies and Toxicities Affecting the Skin
Zinc Deficiency (Parakeratosis)
Etiology
Pigs
Ruminants
Epidemiology
Pigs
Ruminants
Pathogenesis
Clinical Findings
Pigs
Ruminants
Sheep
Infertility in Ewes
Goats
Clinical Pathology
Skin Scraping
Zinc in Serum and Hair
Necropsy Findings
Treatment
Control
Pigs
Ruminants
References
Plant Poisoning Associated With Known Toxins
Dianthrone Derivatives
Fagopyrin
Furocoumarin
Plant Poisonings Associated With Unidentified Toxins
Dermatitis
Photosensitization—Primary; Without Hepatic Lesions
References
Selenium Toxicosis
Etiology
Epidemiology
Occurrence
Pastoral
Dosing Errors
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Farm Risk Factors
Pathogenesis
Clinical Findings
Acute Poisoning
Chronic Poisoning
Clinical Pathology
Necropsy Findings
Acute
Chronic
Samples for Confirmation of Diagnosis
Selenium Levels in Tissue.
Treatment
Control
Further Reading
References
Iodine Toxicosis
Etiology
Clinical Findings
Clinical Pathology/Necropsy
Further Reading
References
Cutaneous Neoplasms
Papilloma and Sarcoid
Squamous-Cell Carcinoma
References
Melanoma
Pathophysiology
Clinical Findings
Clinical Pathology
Treatment
Further Reading
References
Lipoma
References
Mast Cell Tumors
Clinical Findings
Clinical Pathology
Treatment
References
Lymphomatosis
Neurofibromatosis
Histiocytoma
Cutaneous Angiomatosis
Hemangioma and Hemangiosarcoma
Congenital Skin Tumors
Cattle
Pigs
Foals
Further Reading
Congenital and Inherited Defects of the Skin
References
Inherited Albinism
Disorders of Coat Color, Pseudoalbinism, and Lethal Whites
References
Inherited Symmetric Alopecia
Inherited Congenital Hypotrichosis
Viable Hypotrichosis
Nonviable Hypotrichosis
Congenital Hypotrichosis and Anodontia (Anhidrotic Ectodermal Dysplasia)
References
Streaked Hairlessness
Partial Hypotrichosis
Hypotrichosis and Coat-Color Dilution—“Rat-Tail Syndrome” in Calves
Reference
Inherited Hair-Coat-Color-Linked Follicle Dysplasia
Inherited Birthcoat Retention
Inherited Leukoderma
Inherited Epidermal Dysplasia (Baldy Calves)
Inherited Parakeratosis of Calves (Lethal Trait A46, Adema Disease)
Inherited Dyserythropoiesis–Dyskeratosis
Inherited Congenital Absence of the Skin
Epitheliogenesis Imperfecta (Aplasia Cutis)
Reference
Familial Acantholysis
Epidermolysis Bullosa
Junctional Epidermolysis Bullosa (Hereditary Junctional Mechanobullous Disease)
Red Foot Disease of Sheep
Further Reading
References
Inherited Hyperbilirubinemia and Photosensitization
Inherited Congenital Ichthyosis (Fish-Scale Diseases)
Inherited Dermatosis Vegetans of Pigs
Dermatosparaxis (Hyperelastosis Cutis)
References
Inherited Melanoma
Inherited Hyperhidrosis
Lavender Foal Syndrome
Hereditary Equine Regional Dermal Asthenia (Hyperelastosis Cutis)
References
Dermatosis Vegetans
Further Reading
Pustular Psoriaform Dermatitis (Pityriasis Rosea)
Treatment
Eye and Conjunctival Diseases
Conjunctivitis and Keratoconjunctivitis
Etiology
Specific Conjunctivitis
Cattle
Sheep and Goats
Pigs
Horses
Secondary Diseases in Which Conjunctivitis Is a Significant but Secondary Part of the Syndrome
Cattle
Sheep
Pigs
Horses
Nonspecific Conjunctivitis
Clinical Findings
Clinical Pathology
References
Listerial Keratoconjunctivitis and Uveitis (Silage Eye, Bovine Iritis)
Etiology
Epidemiology
Occurrence
Source of Infection
Environmental Risk Factors
Clinical Findings
Clinical Pathology
Treatment
Control
References
Infectious Bovine Keratoconjunctivitis of Cattle (Pinkeye, Blight)
Etiology
Epidemiology
Occurrence
Source of Infection
Environmental Risk Factors
Transmission
Animal Risk Factors
Immune Mechanisms
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Topical Therapy
Subconjunctival Therapy
Parenteral Therapy
Ancillary Therapy
Control
Vaccination
Further Reading
References
Ovine and Caprine Contagious Ophthalmia (Ovine and Caprine Infectious Keratoconjunctivitis, Contagious Conjunctivo-Keratitis, Pinkeye in Sheep and Goats)
Etiology
Mycoplasma conjunctivae
Other Mycoplasmas
Chlamydophila pecorum
Epidemiology
Occurrence
Source of Infection and Transmission
Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
Further Reading
References
Diseases of the Eyes Associated with Mycoplasma spp.
Thelaziasis (Eyeworm)
References
“Bright Blindness” of Sheep Caused by Bracken Ingestion
Bovine Ocular Squamous-Cell Carcinoma
Etiology
Epidemiology
Occurrence
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
Further Reading
References
Equine Ocular Squamous-Cell Carcinoma
Further Reading
References
Inherited Eye Defects
Combined Ocular Defects
Inherited Nystagmus
Familial Undulatory Nystagmus
Pendular Nystagmus
References
External Ear Diseases
Otitis Externa
Further Reading
Ear-Tip Necrosis
Etiology
Epidemiology
Clinical Signs
Pathology
Differential Diagnosis
Treatment
References
Inherited Crop Ears
17 Metabolic and Endocrine Diseases
Introduction
Metabolic Diseases of Ruminants
Periparturient Period in Cattle and Sheep
Transition Period in Dairy Cows
Voluntary Dry Matter Intake in Periparturient Dairy Cattle
Immunosuppression During the Transition Period
Diseases of Lactation
Relationship Between Lactational Performance and Health of Dairy Cattle
Breed Susceptibility
Management Practices
Occurrence and Incidence of Metabolic Diseases
Record Keeping
Metabolic Profile Testing
Usefulness of Metabolic Profile Testing
Biological and Statistical Basis for Herd Testing
Variables in Dairy-Herd Metabolic Profile Testing
Energy Balance
Nonesterified Fatty Acids
β-Hydroxybutyrate
Glucose
Protein Evaluation
Urea Nitrogen
Albumin
Liver Function and Injury
Macromineral Evaluation
Calcium
Phosphorus
Magnesium
Sodium
Potassium
Hematology
Hematocrit (Packed Cell Volume)
Urine Evaluation
Timing of Blood Tests
In Relation to Feed Changes
In Relation to Feeding
In Relation to Calving Pattern and Seasonal Feeding Changes
Selection of Cows
Dry-Cow Group
Early-Lactation Group
Midlactation Group
Energy.
Protein.
Background Information
Interpretation of Results at the Farm
Written Advice
Milk Production, Activity Meters, and Rumination Monitors
Body-Condition Score
Score: 1
Score: 2
Score: 3
Score: 4
Score: 5
Relationships Among International Body-Condition Scoring Systems
Further Reading
References
Parturient Paresis (Milk Fever)
Etiology
Epidemiology
Occurrence
Cattle
Age.
Breed.
Individual Cows.
Time of Occurrence.
Stressors.
Sheep and Goats
Morbidity and Case Fatality
Clinical Hypocalcemia
Subclinical Hypocalcemia
Risk Factors
Animal Risk Factors
Calcium Homeostasis.
Body-Condition Score.
Dietary and Environmental Risk Factors
Dietary Calcium.
Dietary Potassium.
Dietary Magnesium.
Dietary Phosphorus.
Dietary Cation–Anion Difference.
Economic Importance
Milk Fever Relapses.
Downer-Cow Complications.
Dystocia and Reproductive Disease.
Retained Placenta.
Metritis.
Milk Production.
Mastitis.
Displacement of Abomasum.
Ketosis.
Body Weight.
Culling.
Pathogenesis
Hypocalcemia
Experimental Hypocalcemia
Hypomagnesemia
Hypophosphatemia
Clinical Findings
Cattle
Stage 1
Stage 2
Stage 3
Concurrent Hypomagnesemia.
Sheep and Goats
Clinical Pathology
Serum Muscle Enzyme Activity
Hemogram
Necropsy Findings
Differential Diagnosis
Metabolic Diseases
Diseases Associated With Toxemia and Shock
Injuries to the Pelvis and Pelvic Limbs
Degenerative Myopathy (Ischemic Muscle Necrosis)
Downer-Cow Syndrome
Nonparturient Hypocalcemia
Hypocalcemic Paresis in Sheep and Goats
Hypocalcemia in Sows
Treatment
Standard Treatment
Routes of Administration
IV Route
Typical Response to Intravenous Ca-Borogluconate
Unfavorable Response to Intravenous Ca-Borogluconate
SC Route
Oral Route
Failure to Respond to Treatment
General Management and Clinical Care Procedures
Control
Reduction of Dietary Calcium Available for Intestinal Absorption
Dietary Calcium Concentration in Late Gestation
Practicality of Feeding Diets Low in Calcium
Binding Dietary Calcium
Level of Phosphorus in Diet
Calcium-to-Phosphorus Ratio in Diet
Induction of Mild to Moderate Acidosis During Late Gestation: Cation–Anion Difference
Anionic Salts for Acidification of Prepartum Diets for Dairy Cows
Ammonium Chloride.
Strategies for Supplementing Anion Sources
DCAD and Acid–Base Balance of Dairy Cows on Pasture-Based Diets
Summary of Macromineral Nutritional Strategies for the Prevention of Hypocalcemia in the Soon-to-Calve or Transition Dairy Cow in Pasture-Based Systems
Supplementation of Vitamin D During the Dry Period
Parenteral Vitamin D3 Application
Oral Vitamin D Administration
Oral Calcium Supplementation Around Parturition
Parenteral Calcium Supplementation Around Parturition
Partial Milking
Further Reading
References
Acute Hypokalemia in Cattle
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Skeletal Muscle Potassium Content
Plasma Potassium Concentration
Milk Potassium Concentration
Erythrocyte Potassium Concentration
Urine Potassium Concentration
Salivary Potassium Concentration
Necropsy Findings
Treatment
Control
Further Reading
References
Downer-Cow Syndrome
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Complication of Milk Fever.
Traumatic Injuries to Pelvis and Pelvic Limbs.
Serum Electrolyte Imbalances.
Environmental and Management Risk Factors
Pathogenesis
Compartment Syndrome
Crush Syndrome
Experimental Sternal Recumbency
Clinical Findings
Clinical Examination of the Downer Cow
Clinical Pathology
Necropsy Findings
Treatment
Antiinflammatory Therapy
Fluid and Electrolyte Therapy
Bedding and Clinical Care
Assisted Lifting to Aid Standing
Handling, Transportation, and Disposition of Nonambulatory Cattle
Euthanasia
Control
Further Reading
References
Hypomagnesemic Tetanies
Hypomagnesemic Tetany (Lactation Tetany, Grass Tetany, Grass Staggers, Wheat Pasture Poisoning)
Etiology
Magnesium Homeostasis
Dietary Intake
Renal Excretion
Magnesium Reserves
Lactation
Factors Influencing Absorption of Magnesium
Na : K Ratio in Rumen
Other Factors Influencing Absorption
Magnesium in Pastures and Tetany Hazard
Required Magnesium Concentrations
Magnesium Availability in Pastures and Hazard
Winter Hypomagnesemia
Hypomagnesemia and Hypocalcemia
Summary of Etiology
Epidemiology
Occurrence and Risk Factors for Lactation Tetany
Occurrence and Risk Factors for Wheat (Cereal) Pasture Poisoning
Occurrence and Risk Factors for Winter Hypomagnesemia
Morbidity and Mortality
Pasture Risk Factors
Pasture Species
Cereal Pastures
Season
Fertilization
Soil Type
Animal and Management Risk Factors
Dry Matter Intake
Period of Food Deprivation
Alimentary Sojourn
Climate
Animal Movement
Intensive Dairies
Hypomagnesemia in Sheep
Pathogenesis
Clinical Findings
Acute Lactation Tetany
Subacute Lactation Tetany
Chronic Hypomagnesemia
Parturient Paresis With Hypomagnesemia
Clinical Pathology
Serum Magnesium Concentrations
Magnesium Concentrations in Cerebrospinal Fluid
Vitreous Humor
Urine Magnesium Concentrations
Herd Diagnosis
Necropsy Findings
Treatment
Combined Calcium/Magnesium Therapy
Magnesium Therapy
Provision for Further Cases
Control
Feeding of Magnesium Supplements
Problems With Palatability
Spraying on Hay
Pellets
Routine Daily Drenching
Heavy Magnesium “Bullets”
Top Dressing of Pasture
Foliar Dusting and Spraying
Provision in Drinking Water
Management of Pasture Fields
Provision of Shelter
Time of Calving
Feeding on Hay and Unimproved Pasture
Further Reading
References
Hypomagnesemic Tetany of Calves
Etiology
Magnesium Homeostasis in the Calf
Epidemiology
Occurrence
Risk Factors
Experimental Reproduction
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Supplementary Feeding of Magnesium
Magnesium Alloy Bullets
References
Transport Recumbency of Ruminants
Ketosis and subclinical Ketosis (Hyperketonemia) in Cattle
Etiology
Glucose Metabolism in Cattle
Energy Balance
Hepatic Insufficiency in Ketosis
Ketone Formation
Role of Insulin and Glucagon
Etiology of Bovine Ketosis
Individual Cow Variation
Types of Bovine Ketosis
Primary Ketosis (Production Ketosis)
Secondary Ketosis
Alimentary Ketosis
Starvation Ketosis
Ketosis Resulting From Specific Nutritional Deficiency
Epidemiology
Occurrence
Animal and Management Risk Factors
Age.
Body-Condition Score.
Season.
Other Interactions.
Economic Significance
Pathogenesis
Bovine Ketosis
Clinical Findings
Subclinical Ketosis (Hyperketonemia)
Clinical Pathology
Glucose
Ketones
Milk and Urine Cow-Side Tests
Milk Testing.
Urine Testing.
Milk-Fat-to-Protein Ratio.
Clinical Chemistry and Hematology.
Necropsy Findings
Treatment
Replacement Therapy
Glucose (Dextrose)
Other Sugars
Propylene Glycol and Glycerine/Glycerol
Other Glucose Precursors
Hormonal Therapy
Glucocorticoids.
Miscellaneous Treatments.
Control
Energy Supplements
Propionic Acid and Its Salts
Ionophores
Corticosteroids
Ancillary Agents
General Control
Herd Monitoring.
Further Reading
References
Fatty Liver in Cattle (Fat-Mobilization Syndrome, Fat-Cow Syndrome, Hepatic Lipidosis, Pregnancy Toxemia in Cattle)
Etiology
Epidemiology
Occurrence and Incidence
Risk Factors
Host Factors
Genetics of Lipid Mobilization
Environmental and Dietary Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Serum Biochemistry
Hemogram
Liver Biopsy and Analysis
Ultrasonography of the Liver
Necropsy Findings
Treatment
Fluid and Electrolyte Therapy.
Glucagon.
Glucocorticoids.
Outbreaks in a Herd.
Control
Dry Matter Intake and Energy Balance in the Transition Period
Metabolic Adaptations During the Transition Period
Glucose Metabolism
Lipid Metabolism
Nutritional Management to Support Metabolic Adaptations During the Transition Period
Grouping Strategies
Strategies to Meet Glucose Demands and Decrease NEFA Supply During the Transition Period
Carbohydrate Formulation of the Prepartum Diet.
Direct Supplementation With Glucogenic Precursors.
Added Fat in Transition Diets.
Effects of Specific Fatty Acids on NEFA Supply.
Further Reading
References
Pregnancy Toxemia (Twin Lamb Disease) in Sheep
Etiology
Epidemiology
Primary Pregnancy Toxemia
Fat-Ewe Pregnancy Toxemia
Starvation Pregnancy Toxemia
Secondary Pregnancy Toxemia
Stress-Induced Pregnancy Toxemia
Occurrence
Experimental Reproduction
Animal Risk Factors
Pregnancy
Parity
Breed
Economic Significance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Parenteral Therapy
Oral Therapy
Cesarean Section
Control
Prevention
Further Reading
References
Steatitis, Panniculitis, and Fat Necrosis
References
Inherited Metabolic Diseases of Ruminants
Deficiency of UMP Synthase (Dumps)
References
Hepatic Lipodystrophy in Galloway Calves
Metabolic Diseases of Horses
Equine Pituitary Pars Intermedia Dysfunction (Formerly Equine Cushing Disease)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Diagnostic Confirmation
Necropsy Findings
Treatment
Control
Further Reading
References
Equine Metabolic Syndrome
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Diagnosis
Clinical Pathology
Necropsy
Treatment
Dietary Management
Exercise
Medications
Further Reading
References
Pheochromocytoma (Paraganglioma)
References
Glycogen Branching Enzyme Deficiency in Horses
References
Lactation Tetany of Mares (Eclampsia, Transport Tetany)
References
Equine Hyperlipemia
Etiology
Epidemiology
Occurrence
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Postmortem Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma)
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Thyroid Hormone Assays
Necropsy Findings
Treatment
Control
Further Reading
References
Iodine Deficiency
Etiology
Epidemiology
Occurrence
Risk Factors
Dietary and Environmental Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Inherited Goiter
Diseases Caused by Nutritional Deficiencies
Introduction
Evidence of a Deficiency as the Cause of the Disease
Diet
Abnormal Absorption
Abnormal Utilization of Ingested Nutrients
Abnormal Requirement
Evidence of a Deficiency Associated With the Disease
Evidence Based on Cure or Prevention by Correction of the Deficiency
Monitoring of Nutritional Status
Nutritional Management in Dairy Herds
Levels of Nutritional Service
Level 1: Problem Identification and Analysis
Level 2: Ration Analysis
Level 3: Ration Formulation
Level 4: Total Program Consulting
Nutritional Management of the Beef Breeding Herd
Nutritional Advice for Beef Feedlots
Nutritional Advice for Swine-Herds
Nutritional Advice for Sheep Flocks
Placental and Fetal Growth
Lamb Losses
Ewe Body-Condition Score
Further Reading
Deficiencies of Energy and Protein
Deficiency of Energy
Etiology
Epidemiology
Clinical Findings
Deficiency of Protein
Clinical Findings
Prevention
Further Reading
Reference
Low-Milk-Fat Syndrome
References
Diseases Associated With Deficiencies of Mineral Nutrients
Prevalence and Economic Importance
Diagnostic Strategies
Deficiencies in Developing Countries
Pathophysiology of Trace-Element Deficiency
Laboratory Diagnosis of Mineral Deficiencies
Further Reading
References
18 Diseases Primarily Affecting the Reproductive System
Infectious Diseases Primarily Affecting the Reproductive System
Induction of Parturition
Calves
Lambs
Foals
Piglets
Further Reading
References
Freemartinism in Calves
Further Reading
Buller Steer Syndrome
Etiology
Epidemiology
Occurrence
Risk Factors
Economic Importance
Clinical Findings
Control
Further Reading
Reference
Infectious Diseases Primarily Affecting the Reproductive System
Brucellosis Associated With Brucella Abortus (Bang’s Disease)
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Cattle
Camelids
Wildlife Species
Horses
Pigs and Sheep
Dogs
Methods of Transmission
Parturition/Abortion
Transmission
Spread Between Herds
Spread Between Countries (Breach of Biosecurity)
Congenital Infection
Survival of Organism
Uterine Discharges and Milk
Bulls and Semen
Carrier Cows
Risk Factors
Animal Risk Factors
Management Risk Factors
Pathogen Risk Factors
Immune Mechanisms
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Abortion
Orchitis and Epididymitis
Synovitis
Fistulous Withers
Clinical Pathology
Identification of Brucella spp.
Staining
Culture
Detection by Polymerase Chain Reaction (PCR)
Serologic Tests
Agglutination Tests
Serum Agglutination Test
Rose Bengal Test (Buffered Plate Antigen or Card Test)
Complement Fixation Test
Enzyme-Linked Immunosorbent Assays
Indirect Enzyme-Linked Immunosorbent Assay
Competitive Enzyme-Linked Immunosorbent Assay
Fluorescence Polarization Assay
Brucellin Skin Test
Sensitivity and Specificity of Serologic Tests
Antibodies in Milk
False-Positive Reactors
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control and Eradication
Test and Reduction of Reservoir of Infection
Quarantine
Depopulation
Vaccination
Education
Guidelines
Control by Vaccination
Brucella abortus Strain 19 Vaccine
Efficiency of Brucella abortus Strain 19 Vaccine
Calfhood Vaccination.
Adult Vaccination.
Systemic Reactions to Vaccination With Strain 19
Brucella abortus Strain RB51 Vaccine
Brucella Vaccines in Wildlife
Brucella abortus Strain 19 in Bison
Brucella abortus Strain RB51 in Bison
Brucella abortus Strain RB51 in Elk (Cervus elaphus canadensis)
Control Programs on a Herd Basis
During an Abortion Storm
Heavily Infected Herds in Which Few Abortions Are Occurring
Lightly Infected Herds
Hygienic Measures
Eradication on an Area Basis by Test and Slaughter and Cessation of Calfhood Vaccination
United States
Market Cattle Testing
Milk Ring Testing
Australia
New Zealand
Canada
Further Reading
References
Brucellosis Associated With Brucella Ovis
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Source of Infection
Transmission
Host Risk Factors
Experimental Reproduction
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Semen Examination
Culture
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Eradication
Vaccination
Further Reading
References
Brucellosis Associated With Brucella Suis in Pigs
Etiology
Epidemiology
Geographic Occurrence
Biovar 1
Biovar 2
Biovar 3
Biovar 4
Biovar 5
Host Occurrence
Biovar 1
Biovar 2
Biovar 4
Source of Infection
Host and Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Sows
Boars
Piglets
Clinical Pathology
Culture
Serology
Necropsy Findings
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Control
Vaccination
Test and Disposal
References
Brucellosis Associated With Brucella Melitensis
Etiology
Epidemiology
Geographic Occurrence
Host Occurrence
Source of Infection
Reproductive Tract
Milk
Transmission
In Utero Infection
Colostrum and Milk
Host and Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Culture and Molecular Tests
Serology
Milk Tests
Allergic Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Hygiene
Eradication
Rev. 1 Vaccination
Other Vaccines
Further Reading
References
Abortion in Ewes Associated With Salmonella Abortusovis
Further Reading
Reference
Abortion in Mares and Septicemia in Foals Associated With Salmonella Abortusequi (Abortivoequina) (Equine Paratyphoid)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Chlamydial Abortion (Enzootic Abortion of Ewes, Ovine Enzootic Abortion)
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Experimental Reproduction
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Control
Vaccines
Further Reading
References
Coxiellosis (Q-fever)
Etiology
Epidemiology
Occurrence
Source of Infection and Transmission
Pathogen Risk Factors
Zoonotic Implications
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Control
Further Reading
References
Diseases of the Genital Tract Associated With Mycoplasma Spp.
References
Equine Coital Exanthema
Further Reading
References
Porcine Reproductive and Respiratory Syndrome (PRRS)
Introduction
Etiology
Genome
North American Viruses (Type 2 Porcine Reproductive and Respiratory Syndrome Virus)
European Viruses (Type 1 Porcine Reproductive and Respiratory Syndrome Virus)
High Pathogenicity Viruses
Spread of High Pathogenicity Porcine Reproductive and Respiratory Syndrome
Experimental Infections With High Pathogenicity Porcine Reproductive and Respiratory Syndrome
Economic Loss
Epidemiology
Occurrence
Prevalence of Infection
Morbidity and Mortality
Risk Factors
Animal Risk Factors
Environmental and Management Risk Factors
Pathogen Risk Factors
Methods of Transmission
Presence Within the Herd
Introduction of Vaccinated Animals
Other Sources
Fomites
Meat
Insects
Virus Survival
Economic Importance
Pathogenesis
Effects on Macrophages and Dendritic Cells
Receptors
Virus Entry
Replication
General Effects of Porcine Reproductive and Respiratory Syndrome Virus on the Immune System
Macrophage Damage
Toll-Like Receptors
Modulation of Immune Responses
Cellular Changes (Natural Killer, T-Regulatory, etc.)
Cytokines
Interferons
Differential Effects in Different Parts of the Body
Differential Effects of Different Strains
Strain Variations
High Pathogenicity Porcine Reproductive and Respiratory Syndrome Pathogenesis
Development of Lesions
High Pathogenecity Procine Reproductive and Respiratory Syndrome Responses
Immunology
Clinical Findings
Concurrent Infections
Reproductive Failure
Respiratory Disease
Clinical Signs in High Pathogenicity Porcine Reproductive and Respiratory Syndrome Virus
Clinical Pathology
Acute Phase Proteins
Detection or Isolation of Virus
Boars
Serology
Immunoperoxidase Monolayer Assay Test
Indirect Enzyme-Linked Immunosorbent Assay (iELISA)
Indirect Florescent Antibody Assay (IFAT)
Modified Serum Neutralization Test
Herd Diagnosis
Oral Fluids
Antigen Detection
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Filtration Systems
Eradication of the Virus From the Herd
Depopulation and Repopulation
Control in Infected Herds
Nursery Depopulation
Management of the Gilt Pool
Controlled Infection of Breeding Herd
Control of Secondary Infections
Biosecurity
Vaccine and Vaccination
Vaccination of Gilts
Inactivated Vaccines
Live Vaccines
Vaccination Against High Pathogenicity Porcine Reproductive and Respiratory Syndrome
Vaccination of Boars
Further Reading
References
Menangle
Etiology
Epidemiology
Clinical Signs
Pathology
Diagnosis
Differential Diagnosis
Treatment
Control
Further Reading
References
Japanese Encephalitis (Je; Japanese B Encephalitis)
Etiology
Epidemiology
Pathogenesis
Clinicial Signs
Pathology
Diagnosis
Control
Further Reading
Neosporosis
Etiology
Epidemiology
Occurrence
Methods of Transmission
Experimental Studies
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Dourine (Maladie Du Coit)
Etiology
Epidemiology
Occurrence
Measures of Disease Occurrence
Methods of Transmission
Risk Factors
Immune Mechanisms
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Toxic Agents Primarily Affecting the Reproductive System
Estrogenic Substances
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Farm Risk Factors
Human Risk Factors
Pathogenesis
Clinical Signs
Idiopathic Female Estrogenism
Male Estrogenism
Nymphomania in Cows
Swine Estrogenism
Urethral Obstruction
Clinical Pathology
Necropsy Findings
Further Reading
References
Phytoestrogen Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Factors
Human Factors
Plant Factors
Pathogenesis
Clinical Findings
Ewes
Male Castrates
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Zearalenone Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Farm Risk Factors
Human Risk Factors
Pathogenesis
Clinical Findings
Swine
Ruminants
Horses
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Mare Reproductive Loss Syndrome (Early Fetal Loss, Late Fetal Loss, Fibrinous Pericarditis, and Unilateral Uveitis)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Farm Risk Factors
Pathogenesis
Clinical Findings
Early Fetal Loss
Late Fetal Loss
Fibrinous Pericarditis
Unilateral Uveitis
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Equine Amnionitis and Fetal Loss
References
Plants and Fungi (Unknown Toxins) Affecting the Reproductive System
Plants
Plants Associated With Abortion
Plants Associated With Prolonged Gestation
Plants Associated With Congenital Defects
Fungi
Fungi Associated With Reproductive Dysfunction
Congenital and Inherited Diseases Primarily Affecting the Reproductive System
Chromosomal Translocations in Cattle
References
Inherited Prolonged Gestation (Adenohypophyseal Hypoplasia)
Prolonged Gestation With Fetal Gigantism
Prolonged Gestation With Craniofacial Deformity
Prolonged Gestation With Arthrogryposis
References
Inherited Inguinal Hernia and Cryptorchidism
Inguinal Hernias
Cryptorchidism
References
19 Perinatal Diseases
Introduction
Perinatal and Postnatal Diseases
General Classification
Fetal Diseases
Parturient Diseases
Postnatal Diseases
Perinatal Disease—General Epidemiology
Lambs
Mortality Rates
Major Causes
Fetal Disease
Parturient Disease
Postnatal Disease
Birth Weight
Environmental Factors
Infectious Disease
Other Factors
Recording Systems
Dairy Calves
Mortality Rates
Perinatal Mortality Rates
Neonatal Mortality Rates
Fetal Disease/Abortion
Major Causes
Perinatal Mortality
Postnatal Mortality
Postnatal Disease
Management
Beef Calves
Mortality Rates
Major Causes
Fetal Disease
Parturient Disease
Postnatal Disease
Piglets
Mortality Rates
Major Causes
Fetal Disease
Parturient Disease
Postnatal Disease
Management
Foals
Mortality Rates
Major Causes
Fetal Disease
Parturient Disease
Postnatal Disease
New World Camelid Crias
Mortality Rates
Major Causes
Fetal Disease
Parturient Disease
Postnatal Disease
Further Reading
References
Perinatal Disease—Special Investigation of Any Neonatal Deaths (Illness)
Further Reading
Perinatal Disease—Congenital Defects
Etiology
Chromosomal Abnormality and Inheritance
Viral and Other Infections
Nutritional Deficiency
Poisonous Plants
Farm Chemicals
Physical Insults
Environmental Influences
Epidemiology
Pathogenesis
Structural Malformations and Deformations
Viral Teratogenesis
Inherited Congenital Defects
Clinical and Necropsy Findings
Clinical Pathology
Further Reading
Intrauterine Growth Retardation
Etiology
Further Reading
Physical and Environmental Causes of Perinatal Disease
Perinatology
Heart Rate
Ultrasound Examination
Prematurity
Further Reading
References
Prematurity and Dysmaturity of Foals
References
Parturient Injury and Intrapartum Death
Trauma at Parturition
References
Fetal Hypoxia
References
Hypothermia in Newborns
Lambs
Heat Loss in Excess of Summit Metabolism
Hypothermia From Depleted Energy Reserves
Calves
Piglets
Foals
References
Maternal Nutrition and the Newborn
Further Reading
References
Poor Mother–Young Relationship
References
Teeth Clipping of Piglets
Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)
Normal Transfer of Immunoglobulins
Lactogenic Immunity
Failure of Transfer of Passive Immunity
Determinants of Immunoglobulin Concentration in Colostrum
Volume of Colostrum Ingested
Dairy Cows
Beef Cows
Ewes
Sows
All Species
Efficiency of Absorption
Feeding Methods, “Closure of the Gut,” and Immunoglobulin Absorption
Natural Sucking
Artificial Feeding
Other Influences
Decline of Passive Immunity
Other Benefits of Colostrum
Assessment of Transfer of Passive Immunity
Assessment in the Individual Animal
Assessment Tests on Serum
Radial Immunodiffusion
Lateral-Flow Immunoassay
Turbidimetric Immunoassay
Zinc Sulfate Turbidity Test
Sodium Sulfite Precipitation Test
Serum γ-Glutamyltransferase Activity
Serum Total Protein
Glutaraldehyde Coagulation Test
Latex Agglutination Test
ELISA Snap-Test
Monitoring Colostrum
Brix Refractometry
Specific Gravity
Glutaraldehyde Test
ELISA
Correction of Failure of Transfer of Passive Immunity
Oral Therapy
Parenteral Immunoglobulins
Avoidance of Failure of Transfer of Passive Immunity
Colostrum
Colostrum for Banking
Storage of Colostrum
Pasteurization of Colostrum
Cross-Species Colostrum
Colostrum Supplements
Lacteal-Secretion-Based Preparations
Bovine-Serum-Based Preparations
Administration of Colostrum
Foals
Dairy Calves
Assisted Natural Sucking
Artificial Feeding Systems
Beef Calves
Lambs
Piglets
Further Reading
References
Clinical Assessment and Care of Critically Ill Newborns
Clinical Examination
Sepsis
Prematurity and Dysmaturity
Hypoxia
Hypoglycemia
Endocrine Abnormalities
Diagnostic Imaging
Clinical Pathology
Serum Immunoglobulin Concentration
Hematology
Serum Biochemistry
Blood Gas
Blood Culture
Other Body Fluids
Treatment
Nursing Care
Correction of Failure of Transfer of Passive Immunity
Colostral Immunoglobulin
Critical Plasma IgG Concentrations in Foals
Plasma Transfusion
Nutritional Support
Antimicrobial Treatment
Fluid Therapy
Respiratory Support
Gastroduodenal Ulcer Prophylaxis
Common Complications
Prognosis
Further Reading
References
Stillbirth/Perinatal Weak-Calf Syndrome
Historical Aspects
Dummy-Calf Syndrome
Etiology and Epidemiology
Fetal Infections
Maternal Nutritional Deficiency Causing Fetal Underdevelopment
Placental Insufficiency
Fetal Hypoxemia
Dystocia and Traumatic Injuries at Birth
Premature Expulsion of Placenta
Necropsy Findings
Treatment
Control and Prevention
Further Reading
References
Diseases of Cloned Offspring
Further Reading
References
Equine Neonatal Maladjustment Syndrome (Neonatal Encephalopathy, Dummy Foal, Barkers, and Wanderers)
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Clinical Pathology
Diagnostic Confirmation
Necropsy Findings
Samples for Postmortem Diagnostic Confirmation
Treatment
Control
Further Reading
References
Neonatal Infectious Diseases
Etiology
Calves
Pigs
Foals
Lambs
Calves
Lambs
Foals
All Species
Epidemiology
Sources of Infection
Postnatal Infection
Prenatal Infection
Routes of Transmission
Risk Factors and Modulation of Infection
Immunity
Immune Responsiveness
Exposure Pressure
Age at Exposure
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Control
References
Principles of Control and Prevention of Neonatal Infectious Diseases
Reduction of Risk of Acquisition of Infection From the Environment
Removal of the Newborn From the Infectious Environment
Increasing the Nonspecific Resistance of the Newborn
Increasing the Specific Resistance of the Newborn
Further Reading
References
Colibacillosis of Newborn Calves, Piglets, Lambs, Kids, and Foals
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Cattle and Calves
Piglets
Morbidity and Mortality Rates
Calves
Piglets
Risk Factors
Animal Risk Factors
Animal Species
Age and Birth Weight
Immunity and Colostrum
Environmental and Management Risk Factors
Meteorologic Influences
Nutrition and Feeding Methods
Standard of Housing and Hygiene
Source of the Organism and Its Ecology and Transmission
Pathogen Risk Factors
Virulence Factors of E. coli
Lambs
Zoonotic Implications of E. coli
Pathogenesis
Septicemic Colibacillosis (Coliform Septicemia)
Enteric Colibacillosis
Enterotoxigenic Colibacillosis
Diarrhea, Dehydration, Metabolic Acidosis, and Electrolyte Imbalance
Hyperkalemia in the Diarrheic Calf
Hypernatremia in the Diarrheic Calf
Effect of Colostral Immunoglobulin Status
Intestinal Mucosa
Attaching and Effacing Colibacillosis
Synergism Between Enteropathogens
Summary of Pathogenesis
Clinical Findings
Calves
Coliform Septicemia
Predictors of Septicemia
Enteric Colibacillosis
Lambs and Goat Kids
Piglets
Coliform Septicemia
Enterotoxigenic Colibacillosis
Newborn Piglet Diarrhea
Postweaning Diarrhea
Clinical Pathology
Culture and Detection of Organism
Hematology and Serum Biochemistry
Necropsy Findings
Samples for Confirmation of Diagnosis
Coliform Septicemia
Enteric Colibacillosis
Treatment
Coliform Septicemia
Enteric Colibacillosis
Fluid and Electrolyte Replacement
Categorizing Diarrheic Calves Into Treatment Groups
Antimicrobial Therapy
Change in Small Intestinal Bacterial Flora in Calves With Diarrhea
Incidence of Bacteremia in Calves With Diarrhea
Antimicrobial Susceptibility of Fecal Escherichia coli Isolates
Surveillance for Antimicrobial Resistance in Escherichia coli Isolates
Antimicrobial Susceptibility of Blood Escherichia coli Isolates
Evidence-Based Recommendations for Antimicrobial Treatment of Diarrheic Calves
Administration of Oral Antimicrobials to Treat Escherichia coli Overgrowth of the Small Intestine
Administration of Parenteral Antimicrobials to Treat Escherichia coli Bacteremia
Use of Antimicrobials That Are of Critical Importance for Human and Veterinary Medicine
Immunoglobulin Therapy
Analgesic and Antiinflammatory Therapy
Antimotility Drugs
Intestinal Protectants
Alteration of the Diet
Probiotics
Clinical Management of Outbreaks
Control
Reduction of the Degree of Exposure of the Newborn to the Infectious Agents
Dairy Calves
Beef Calves
Veal Calves
Lambs and Kids
Piglets
Provision of Maximum Nonspecific Resistance With Adequate Colostrum and Optimum Animal Management
Colostrum Management
Quality of Colostrum
Specific Gravity
Frozen and Thawed Colostrum
Pasteurization of Colostrum
Colostrum Supplements and Replacers
Purified Bovine Immunoglobulin
Beef Calves
Lambs
Piglets
Increasing Specific Resistance of the Newborn by Vaccinating the Pregnant Dam or the Newborn
Calves
Piglets
Competitive Exclusion Culture
Lambs and Kids
Further Reading
References
Watery Mouth of Lambs (Rattle Belly, Slavers)
Etiology
Epidemiology
Occurrence
Animal and Environmental Risk Factors
Experimental Disease
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Omphalitis, Omphalophlebitis, and Urachitis in Newborn Farm Animals (Navel Ill)
Omphalitis
Omphalophlebitis
Omphaloarteritis
Urachitis
Control
References
Neonatal Streptococcal Infection
Etiology
Epidemiology
Occurrence and Prevalence
Source of Infection
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Foals
Piglets
Lambs
Calves
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Neonatal Neoplasia
20 Diseases of the Mammary Gland
Introduction
Bovine Mastitis
General Features
Contagious Mastitis Pathogens
Teat Skin Opportunistic Mastitis Pathogens
Environmental Mastitis Pathogens
Etiology
Major Pathogens
Contagious Pathogens
Environmental Pathogens
Minor Pathogens
Uncommon Mastitis Pathogens
Epidemiology
Occurrence and Prevalence of Infection
Prevalence
Incidence
Relative Prevalence of Infection With Intramammary Pathogens
Contagious Pathogens
Teat Skin Opportunistic Pathogens
Environmental Pathogens
Heifers
Distribution of Pathogens in Clinical Mastitis
Source of Infection
Contagious Pathogens
Teat Skin Opportunistic Pathogens
Environmental Pathogens
Methods of Transmission
Risk Factors
Animal Risk Factors
Age and Parity
Stage of Lactation
Season of Year
Somatic Cell Count
Breed
Milking Characteristics and Morphology of Udder and Teat
Physical Condition of Teat
Udder Hygiene
Nutritional Status
Genetic Resistance to Mastitis
Somatic Cell Count.
Milk Yield
Other Concurrent Diseases
Immunologic Function of Mammary Gland
Previous Mastitis
Preexisting Intramammary Infections
Use of Recombinant Bovine Somatotropin
Environmental and Management Risk Factors
Quality and Management of Housing
Milking Practices
Pathogen Risk Factors
Viability of Pathogens
Virulence Factors
Colonizing Ability
Toxins
Production and Economic Losses
Subclinical Mastitis
Clinical Mastitis
Zoonotic Potential
Pathogenesis
Clinical Findings
Abnormal Secretion
Abnormal Gland
Abnormal Cow (Systemic Response)
Further Reading
References
Diagnosis of Bovine Mastitis
Detection of Clinical Mastitis
Detection of Subclinical Mastitis
Detection at the Herd Level
Bulk Tank Milk Somatic Cell Counts
Culture of Bulk Tank Milk
Detection at the Individual Cow Level
Culture of Individual Cow Milk
Indirect Tests for Subclinical Mastitis
The Somatic Cell Count of Composite or Quarter Samples
California Mastitis Test of Quarter Samples
NAGase Test of Composite or Quarter Samples
Electrical Conductivity Tests of Quarter Samples
Comparison of Indirect Methods
Hematology and Serum Biochemistry
Ultrasonography of the Mammary Gland
Infrared Thermography of the Mammary Gland
Biopsy of Mammary Tissue
Necropsy Findings
Further Reading
References
Treatment of Bovine Mastitis
Historical Aspects of Antimicrobial Therapy for Clinical and Subclinical Mastitis
Treatment Strategy
Is Antimicrobial Therapy Indicated?
Type of Pathogen Involved
Type and Severity of the Inflammatory Response
Duration of Infection
Stage of Lactation
Age and Pregnancy Status of Cow
Which Route of Administration (Intramammary, Parenteral, or Both?)
Which Antimicrobial Agent Should Be Administered?
What Should Be the Frequency and Duration of Treatment?
Intramammary Antimicrobial Therapy
Infusion Procedure
Parenteral Antimicrobial Therapy
Treatment of Lactating Quarters
Emergency Treatment When the Type of Infection Is Unknown
Treatment When the Infecting Organism Is Known
Treatment of Subclinical Mastitis
Antiinflammatory Agents
Supportive Therapy
Adjunctive Therapy
Magnitude of Response to Therapy
Dry Cow Therapy
Prepartum Antimicrobial Therapy in Heifers
Treatment of Mastitis on Organic Dairy Farms
Antimicrobial Residues in Milk and Withholding Times
Antimicrobial Residue Tests
Permanently Drying Off Chronically Affected Quarters
Further Reading
References
Mastitis Pathogens of Cattle
Mastitis of Cattle Associated With Common Contagious Pathogens
Staphylococcus aureus
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Coagulase-Positive Staphylococci
Source of Infection and Method of Transmission
Risk Factors
Animal Risk Factors
Local Defense Mechanisms
Colonization With Minor Pathogens
Parity of Cow
Presence of Other Diseases
Heredity
Immune System
Environmental and Management Risk Factors
Pathogen Risk Factors
Virulence Factors
Genotype of Strains
Methicillin-Resistant Staphylococcus Aureus
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Chronic Staphylococcus aureus Mastitis
Acute and Peracute Staphylococcus aureus Mastitis
Clinical Pathology
Culture of Individual Cow Milk
Culture of Bulk Tank Milk
Somatic Cell Counts and California Mastitis Test
Enzyme-Linked Immunosorbent Assays for Antibody in Milk
Acriflavine Disk Assay
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Inadequate Penetration of Antimicrobial Agent
Antimicrobial Resistance
Lactating Cow Therapy
Peracute Mastitis
Dry Cow Therapy
Control
Vaccination
Further Reading
References
Streptococcus agalactiae
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Source of Infection
Transmission of Infection
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Culture From Bulk Tank Milk Samples
Total Bacterial Count
Culture From Individual Cow Samples
Somatic Cell Count
Necropsy Findings
Treatment
Blitz Therapy
Control
Vaccination
Biosecurity
Further Reading
References
Corynebacterium bovis
Etiology
Epidemiology
Clinical Findings
Treatment
Control
References
M. bovis and Other Mycoplasma sp.
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Source of Infection
Transmission
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Mastitis of Cattle Associated With Teat Skin Opportunistic Pathogens
Coagulase-Negative Staphylococci
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Treatment
Control
Further Reading
References
Mastitis of Cattle Associated With Common Environmental Pathogens
Coliform Mastitis Associated With Escherichia coli, Klebsiella sp., and Enterobacter aerogenes
Etiology
Epidemiology
Occurrence of Clinical Mastitis
Prevalence of Infection
Duration of Infection
Source of Infection and Mode of Transmission
Morbidity and Case Fatality
Risk Factors
Pathogen Risk Factors
Environmental Risk Factors
Bedding
Animal Risk Factors
Somatic Cell Count
Neutrophil Recruitment and Function
Selenium and Vitamin E Status
Stage of Lactation and Defense Mechanism
Contamination of Teat Duct
Downer Cows
Other Defense Factors
Lactoferrin and Citrate.
Serum Antibody to E. coli.
Pathogenesis
Stage of Lactation
Neutrophil Response
Numbers of Bacteria in Milk
Experimental Endotoxin-Induced Mastitis
Clinical Findings
Accuracy of Clinical Diagnosis
Clinical Pathology
Culture of Milk
Somatic Cell Count and California Mastitis Test Scores
Hematology
Endotoxin Presence in Milk and Plasma
Serum Biochemistry
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Treatment Trials Using Antimicrobial Agents and Untreated Controls
Parenteral Antimicrobial Agents
Intramammary Antimicrobial Agents
Stripping of the Affected Quarter
Fluid and Electrolyte Therapy
Antiinflammatory Agents
Combination Therapy
Control
Management of Outbreaks
Housing and Environment
Bedding
Regular Daily Cleaning of Barns
Milking Procedures
Premilking Teat Disinfection
Postmilking Barrier Teat Dips
Nutrition
Prevention of Infection During Dry Period
Recumbent Cows
Milking Machine
Vaccination
Core Lipopolysaccharide Antigen Vaccine
Further Reading
References
Environmental Streptococci
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Source of Infection
Risk Factors
Environmental Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Treatment
Antimicrobial Agents
Control
Vaccination
References
Trueperella pyogenes
Etiology
Epidemiology
Occurrence and Prevalence of Infection
Source of Infection and Mode of Transmission
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Mastitis of Cattle Associated With Less Common Pathogens
Pseudomonas aeruginosa
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
References
Mannheimia (Pasteurella) species
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Nocardia sp.
Etiology
Epidemiology
Occurrence
Source of Infection and Mode of Transmission
Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Bacillus sp.
Campylobacter jejuni
Clostridium perfringens type A
Fusobacterium necrophorum
Histophilus somni
Listeria monocytogenes
Mycobacterium sp.
Serratia sp.
Fungi and Yeasts
Algae
Traumatic Mastitis
References
Control of Bovine Mastitis
Udder Health Improvement
Economic Benefits, Incentives, and Penalties
Requirements
Options in the Control of Mastitis
Eradication
Decreasing the Infection Rate
Legislative Control
A Voluntary Program
Principles of Controlling Bovine Mastitis
Dynamics of Infection
1. Eliminate Existing Infections
2. Prevent New Infections
3. Monitor Udder Health Status
Mastitis Control Programs
Ten-Point Mastitis Control Program
1. Udder Hygiene and Proper Milking Methods
Establish and Maintain a Regular Milking Schedule in a Stress-Free Environment
Ensure That Teats Are Clean and Dry Before Milking
Udder Hygiene Score
Premilking Cow Preparation
Check Foremilk and Udder for Mastitis
Premilking Teat Disinfection
Attach the Milking Unit Properly
Minimize Machine Stripping and Avoid Liner Slips
Avoid Overmilking or Removing the Unit Under Vacuum
Use an Effective and Safe Postmilking Teat Germicide (Teat Dip) After Every Milking
Iodine Formulations
Chlorhexidine
Linear Dodecyl Benzene Sulfonic Acid Products
Quaternary Ammonium Compounds
Sodium Hypochlorite
External Teat Sealants (Barrier Teat Dips)
Selection and Use of Teat Disinfectants
Establish Milking Order and Segregation Programs
Disinfect Teat Liners
2. Proper Installation, Function, and Maintenance of Milking Equipment
Milking System Function and Objectives
Components of a Milking System
Vacuum System
Vacuum Pump
Vacuum Reserve Tank
Vacuum Regulator
Pulsation System
Milk Transport System
Milk Claw
Milk Pipeline
Bulk Milk Tank
Relationship of Milking Equipment to Udder Health
Maintenance and Evaluation of Milking Equipment
Robotic Milking
3. Dry Cow Management and Therapy
Epidemiology of Intramammary Infection During the Dry Period
Incidence of New Infections
Types of Pathogen Causing New Infections During the Dry Period
Risk Factors That Affect Susceptibility in Dry Cows
Teat-End Protection
Resistance Mechanisms Within the Mammary Gland
Milk Production at Dry Off
Method of Drying Off
Parity
Risk Factors That Affect Susceptibility in Heifers
Udder Health Management Strategies for Dry Cows
Antimicrobial Therapy (Dry Cow Therapy)
Blanket Versus Selective Dry Cow Therapy
Factors Affecting the Success of Antimicrobial Treatment of Dry Cows
Internal Teat Sealants
External Teat Sealants
Teat Disinfection
Intramammary Devices
Vaccination of the Dry Cow
Management of the Environment for Dry Cows
Nutritional Management of Dry Cows
4. Appropriate Therapy of Mastitis During Lactation
5. Culling Chronically Infected Cows
Biosecurity for Herd Replacements
Know the Farm of Origin
Know the Cows
Protect the Home Herd
6. Maintenance of an Appropriate Environment
Global Environmental Influences
Classification of Environmental Influences
External Environmental Influences on Mastitis Control
Regional Environment
Local Environment
Internal Environmental Influences on Mastitis Control
Housing
Nutritional Management
Management Approach
General Hygiene
Udder Singeing
Use of Recombinant Bovine Somatotropin
Environmental Control in an Udder Health Management Program
7. Good Record Keeping
Objectives and Uses of Clinical Mastitis Records
Recording Clinical Mastitis Data
Using Clinical Mastitis Monitoring Systems
Cow Versus Herd Clinical Mastitis Problems
Probability of Recurrence of Clinical Mastitis in the Same Lactation
Stage of Lactation and Seasonal Profile
Days of Discarded Milk
8. Monitoring Udder Health Status
Monitoring Udder Health at the Herd Level
Bulk Tank Milk Somatic Cell Counts
Herd Average of Weighted Individual Cow Somatic Cell Count
Other Herd-Level Somatic Cell Count Monitors
Herd Average Somatic Cell Score
Percentage of Herd Over Somatic Cell Count Threshold
Percentage of Herd Changing Somatic Cell Count to Over Threshold
Bacteriologic Culture of Bulk Milk
Herd-Level Measures of Clinical Mastitis
Monitoring Udder Health of Individual Cows
Bacteriologic Culture of Milk
Somatic Cell Counts
Problem Solving Using Individual Cow Somatic Cell Counts
9. Periodic Review of the Udder Health Management Program
10. Setting Goals for Udder Health Status
Relationship of Udder Health to Productivity and Profitability
Production Losses From Increased Somatic Cell Count
Clinical Mastitis and Lost Productivity
Assessment of the Cost-Effectiveness of Mastitis Control
Further Reading
References
Miscellaneous Abnormalities of the Teats and Udder
Lesions of the Bovine Teat
Reference
Lesions of the Bovine Teat and Udder
Bovine Herpes Mammillitis
Etiology
Epidemiology
Occurrence
Origin of Infection and Transmission
Animal and Pathogen Risk Factors
Economic Importance
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology and Necropsy Findings
Diagnosis
Treatment
Control
References
Lesions of the Bovine Udder Other Than Mastitis
Udder Impetigo
Udder Cleft Dermatitis
Blood in the Milk (Mammary Gland Hematoma)
Udder Edema
Rupture of the Suspensory Ligaments of the Udder
Agalactia
Milk Drop Syndrome
Low Milk Fat Syndrome
References
“Free” Or “Stray” Electricity as a Cause of Failure of Letdown
Neoplasms of the Udder
Teat and Udder Congenital Defects
Further Reading
References
Milk Allergy
Mastitis of Sheep
Etiology
Epidemiology
Occurrence
Staphylococcal Mastitis
Mannheimia Mastitis
Streptococcal Mastitis
Pathogenesis
Clinical Findings
Staphylococcal Mastitis
Mannheimia Mastitis
Clostridial Mastitis
Caseous Lymphadenitis and Mastitis
Pseudomonal Mastitis
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Staphylococcal Mastitis
Mannheimia Mastitis
Further Reading
References
Mastitis of Goats
Staphylococcal Mastitis
Streptococcal Mastitis
Pseudomonas Mastitis
Summer Mastitis
Other Infections
Clinical Findings
Treatment and Control
References
Contagious Agalactia in Goats and Sheep
Etiology
Epidemiology
Occurrence
Prevalence
Transmission
Experimental Reproduction
Host Risk Factors
Pathogen Risk Factors
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Mastitis of Mares
References
Postpartum Dysgalactia Syndrome of Sows
Etiology
Mastitis
Epidemiology
Occurrence
Morbidity and Case Fatality
Risk Factors
Feed
Housing
Management
Animal Factors
Microorganisms
Pathogenesis
Clinical Findings
Clinical Pathology
Examination of Milk
Hematology and Serum Biochemistry
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Supplementation of Piglets
Control
Further Reading
References
21 Systemic and Multi-Organ Diseases
Diseases of Complex or Undetermined Etiology
Cold Cow Syndrome
Recumbency in horses of undetermined etiology
Epidemiology
Examination of the Recumbent Horse
History
Physical Examination
Management and Care
Complications—Prevention
Further Reading
References
Thin Sow Syndrome
Etiology
Epidemiology
Clinical Findings
Control
Feeding During Pregnancy
Feeding During Lactation
Wild Boar as Vectors for Infectious Disease
Risk Factors
Disease in General
African Swine Fever
Astroviruses
Aujesky’s Disease
Brucellosis
Campylobacter Spp.
Chlamydiae
Classical Swine Fever
Cryptosporidia
Escherichia Coli
Erysipelas
Fasciola Hepatica
Foot-and-Mouth Disease
Hepatitis E
Leptospira Spp.
Lymphadenitis
Mycobacterium Bovis
Mycoplasma Hyopneumoniae
Parasites
Porcine Circovirus Type 2
Porcine Parvovirus
Porcine Reproductive and Respiratory Syndrome
Porcine Sapelovirus
Salmonella Serotypes
Swine Influenza
Toxoplasma
Trichinella
Control
References
Multi-Organ Diseases Due to Bacterial Infection
Anthrax
Etiology
Epidemiology
Occurrence
Source of the Infection
Transmission of the Infection
Risk Factors
Host Risk Factors
Pathogen Risk Factors
Environment Risk Factors
Economic Importance
Zoonotic Potential
Pathogenesis
Clinical Findings
Cattle and Sheep
Pigs
Horses
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Immunization
Further Reading
References
Bovine Tuberculosis
Etiology
Epidemiology
Occurrence
Source of Infection
Cattle
Wildlife Reservoirs
Methods of Transmission
Ingestion
Other Routes
Risk Factors
Environment Risk Factors
Host Risk Factors
Pathogen Risk Factors
Economic Importance
Zoonotic Importance
Pathogenesis
Clinical Findings
Cattle
Lungs
Intestine
Uterus
Mastitis
Pigs
Horses
Sheep and Goats
New World Camelids
Clinical Pathology
Direct Tests
Microscopic Examination
Culture
Nucleic Acid Recognition Methods
Indirect Tests
Intradermal Tuberculin Test
Special Aspects of Sensitivity to Tuberculin
Summary of Testing Procedures in Cattle
Tuberculin Testing in Other Species
Pigs
Horses
Sheep and Goats
New World Camelids
Interferon-γ Assay (IFN-γ)
Lymphocyte Proliferation Test
Serologic Tests for Diagnosis of Tuberculosis
Necropsy Findings
Cattle, Sheep, and Goats
Pigs
Horses
Samples for Confirmation of Diagnosis
Treatment
Control
Control on a Herd Basis
Tuberculin Testing
Retesting
Prevention of Spread
Control on an Area Basis
Education
Staging
Vaccination
Test and Slaughter
Problems in Tuberculosis Eradication
No-Visible-Lesion Reactors
“Breakdowns”
“Traceback”
Large Herds
Wildlife Reservoirs
Control of Tuberculosis in Pigs
Further Reading
References
Tuberculosis Associated With Mycobacterium tuberculosis
References
Mycobacteriosis Associated With Mycobacterium avium intracellulare Complex and With Atypical Mycobacteria
Etiology
Epidemiology
Occurrence
Source and Transmission
Economic Importance
Zoonotic Importance
Clinical and Necropsy Findings
Cattle
Goats and Sheep
Deer
Horses
Pigs
Clinical Pathology
Tuberculin Testing
Treatment and Control
Further Reading
References
Yersiniosis
Etiology
Epidemiology
Occurrence
Yersinia pseudotuberculosis
Yersinia enterocolitica
Zoonotic Implications
Yersinia pseudotuberculosis
Yersinia enterocolitica
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
Tularemia
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Transmission
Pathogen Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Sheep
Pigs
Horses
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Melioidosis
Etiology
Epidemiology
Occurrence
Risk Factors
Source and Methods of Transmission
Pathogen Risk Factors
Experimental Production
Zoonotic Implications
Pathogenesis
Clinical Findings
Sheep
Goats
Pigs
Horses
Clinical Pathology
Necropsy
Diagnostic Confirmation
Treatment
Control
Further Reading
References
Heartwater (COWDRIOSIS)
Etiology
Epidemiology
Occurrence
Measures of Disease Occurrence
Method of Transmission
Risk Factors and Immune Mechanisms
Economic Importance
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Histophilus Septicemia of Cattle (Histophilus somni or Haemophilus somnus Disease Complex)
Etiology
Epidemiology
Prevalence of Infection
Occurrence of Disease
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Adherence.
Lipooligosaccharides (LOS or Endotoxin).
Antigen Variation of Surface Proteins.
Immunoglobulin-Binding Proteins.
Transferrin-Binding Proteins.
Biofilm Synthesis.
Methods of Transmission
Immune Mechanisms
Pathogenesis
Clinical Findings
Thrombotic Meningoencephalitis (TME)
Respiratory Disease
Myocarditis
Clinical Pathology
Hematology
Culture of Organism
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Metaphylactic Antimicrobial Therapy
Vaccination
Further Reading
References
Septicemia and Thrombotic Meningoencephalitis in Sheep Associated With Histophilus Somni
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
Tick Pyemia of Lambs (Enzootic Staphylococcosis of Lambs)
Etiology
Epidemiology
Pathogenesis
Clinical and Necropsy Findings
Treatment and Control
Further Reading
Reference
Septicemic Pasteurellosis of Cattle (Hemorrhagic Septicemia)
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Environmental Risk Factors
Transmission
Pathogenesis
Clinical Findings
Clinical Pathology
Culture and Detection of Bacteria
Serology
Necropsy Findings
Treatment
Control
Antimicrobial Metaphylaxis
Vaccines
Further Reading
References
Pasteurellosis of Sheep and Goats
Septicemic Pasteurellosis of Suckling Lambs
Mannheimia mastitis in sheep
Pneumonic Pasteurellosis Affecting Wildlife
References
Pasteurellosis of Swine
Pneumonic Pasteurellosis
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Necropsy Findings
Diagnosis
Treatment
Control
Septicemic Pasteurellosis
Samples for Confirmation of Diagnosis
References
Streptococcus suis Infection of Young Pigs
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Environmental and Management Factors
Pathogen Risk Factors
Capsules
Proteins
Suilysin
Hemolysin
Other Properties
Adhesion
Zoonotic Implications
Pathogenesis
Clinical Findings
Clinical Pathology
Culture or Detection of Organism
Serology
Necropsy Findings
Samples for Confirmation of Diagnosis
Diagnosis
Treatment
Antimicrobials
Control
Environment and Management
Mass Medication of Feed
Vaccination
References
Streptococcal Lymphadenitis of Swine (Jowl Abscesses, Cervical Abscesses)
Erysipelas in Swine
Etiology
Epidemiology
Occurrence
Prevalence of Infection
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Zoonotic Implications
Pathogenesis
Clinical Findings
Hyperacute Form
Acute and Subacute Forms
Chronic Form
Clinical Pathology
Detection of Organism
Hematology
Serology
Necropsy Findings
Acute and Subacute Forms
Chronic Form
Diagnosis
Samples for Confirmation of Diagnosis
Treatment
Antimicrobial Therapy
Control
Eradication
Immunization
Antierysipelas Serum
Vaccination
Vaccination Program
References
Actinobacillus Septicemia in Piglets
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Necropsy Findings
Diagnosis
Treatment
References
Klebsiella Pneumoniae Septicemia in Pigs
Chlamydial Infection in the Pig
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Necropsy Findings
Diagnosis
Treatment
Control
References
Multi-Organ Diseases Due to Viral Infection
Foot-and-Mouth Disease (Aphthous Fever)
Etiology
Epidemiology
Occurrence
Prevalence
Morbidity and Case-Fatality Rate
Methods of Transmission
Risk Factors
Host Factors
Environmental and Pathogen Factors
Immune Mechanism
Experimental Reproduction
Economic Importance
Zoonotic Implications
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Control by Eradication
Vaccination
Further Reading
References
Rift Valley Fever
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Source of Infection
Method of Transmission
Experimental Reproduction
Zoonotic Implications
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Vaccines
Further Reading
References
Bluetongue
Etiology
Epidemiology
Occurrence
Host Occurrence
Method of Transmission
Culicoides Species
Other Vectors
Overwintering
Transplacental Infection
Persistent Infection
Venereal Transmission
Oral Transmission
Pathogen and Vector Risk Factors
Climate
Serotype Occurrence
Host Risk Factors
Cattle
Sheep
Goats and Wild Ruminants
New World Camelids
Morbidity and Case Fatality
Experimental Reproduction
Economic Importance
Pathogenesis
Sheep
Cattle and Wild Ruminants
Clinical Findings
Sheep
Cattle
Goats
New World Camelids
Wild Ruminants
Clinical Pathology
Virus Isolation
Detection of Antigen or Nucleic Acid
Serologic Tests
Necropsy Findings
Sheep
Cattle
Samples for Confirmation of Diagnosis
Treatment
Control
Reduction of Infection Through Vector Abatement
Vaccination
International Movement of Livestock
Further Reading
References
Malignant Catarrhal Fever (Bovine Malignant Catarrh, Malignant Head Catarrh)
Etiology
Epidemiology
Occurrence and Prevalence
Alcelaphine MCF
Sheep-Associated MCF
Methods of Transmission
Alcelaphine MCF
Sheep-Associated MCF
Experimental Reproduction
Environment Risk Factors
Animal Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Head-and-Eye Form
Peracute and Alimentary Tract Forms
Mild Form
Pigs
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Malignant Catarrh in Pigs
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Pathology
Diagnosis
References
Jembrana Disease
Etiology
Epidemiology
Occurrence
Transmission
Experimental Reproduction
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment and Control
Further Reading
References
Bovine Ephemeral Fever
Etiology
Epidemiology
Occurrence
Source of Infection
Method of Transmission
Experimental Reproduction
Environment Risk Factors
Animal Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Nairobi Sheep Disease
Further Reading
References
Wesselsbron Disease
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Source of Infection and Method of Transmission
Zoonotic Implications
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
References
Caprine Herpesvirus-1 Infection
Etiology
Epidemiology
Occurrence
Transmission and Experimental Reproduction
Economic Importance
Pathogenesis
Clinical Signs
Adults
Newborn Kids
Clinical Pathology
Necropsy Findings
Adults
Newborn Kids
Samples for Confirmation of Diagnosis
Treatment and Control
References
Equine Viral Arteritis
Etiology
Epidemiology
Occurrence
Origin of Infection and Transmission
Horizontal Transmission
Venereal Transmission
Immunity
Animal Risk Factors
Economic Importance
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment and Control
Further Reading
References
African Horse Sickness
Etiology
Epidemiology
Occurrence
South Africa
Transmission of Infection
Zebra
Midges
Risk Factors
Environment Factors
Animal Factors
Economic Importance
Zoonotic Disease
Pathogenesis
Clinical Findings
Acute (Pulmonary) Horse Sickness (Dunkop)
Subacute (Cardiac) Horse Sickness (Dikkop)
Horse Sickness Fever
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Prevention of Introduction
Slaughter of Sick or Viremic Animals
Reduce Exposure to Biting Midges
Vaccination
References
Equine Encephalosis
Etiology
Epidemiology
Clinical Signs
Clinical Pathology
Necropsy Findings
References
Getah Virus Infection
Etiology
Epidemiology
Clinical Signs
Necropsy Findings
Treatment
Control
References
Classical Swine Fever (Hog Cholera)
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Animal Risk Factors
Pathogen Risk Factors
Virulence Characteristics
Resistance of Virus
Immune Mechanisms
Economic Importance
Pathogenesis
Clinical Findings
Diagnosis
Peracute and Acute Disease
Nervous Manifestations
Chronic Disease
Reproductive Failure
Clinical Pathology
Hematology
Diagnostic Tests
Detection of Virus
Fluorescent Antibody Techniques
Antigen-Capture ELISA
Agar Gel Precipitation Test
Differentiation of Swine Fever Virus From Other Pestiviruses
PCR Tests
Serologic Tests
Samples for Laboratory
Necropsy Findings
Diagnosis
Detection of Antigen
Samples for Confirmation of Diagnosis
Serology
Treatment
Control
Control of Outbreaks in Hog Cholera–Free Areas
Control Where Hog Cholera Is Endemic
Hog Cholera Eradication
Immunization Methods
Serum–Virus Vaccination
Attenuated Vaccines
Inactivated Vaccines
Further Reading
References
African Swine Fever
Etiology
Epidemiology
Geographic Occurrence
Africa
Europe
The Recent Disease Outbreak
Species Affected
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Pathogen Factors
Immune Mechanisms
Pathogenesis
Clinical Findings
Clinical Pathology
Hematology
Diagnosis
Detection of the Virus
Serologic Tests
Necropsy Findings
Samples for Confirmation of Diagnosis
Serology
Treatment
Control
Vaccines
Further Reading
References
Porcine Circovirus–Associated Disease
Etiology
Genome Studies
Transcription
Embryo Infectivity
Virus Import
Replication
Genotypes
Epidemiology
Wild Boar
Prevalence
Environmental Survival
Transmission
Horizontal Transmission
Vertical Transmission
Risk Factors
Occurrence
Breeding
Concurrent Infections
Pathogenesis
Host–Virus Interactions
Cytokine Studies
Interferons
Immunity
Necropsy Findings
Gross Pathology
Fetal Pathology
Pathology in Piglets and Pigs
Histopathology
Porcine Dermatitis and Nephropathy Syndrome
Ultrastructural Changes
Clinical Signs
Clinical Pathology
Diagnosis
Serology
Diagnosis in Boars
Virus Detection
Detection of Viral Antigen by Immunohistochemistry
Oral Fluids
Control
Vaccination
The Vaccines
Vaccine Effects
Sow Compared with Piglet Vaccination
Sow Vaccination
Semen Shedding in Boars
Piglet Vaccination
Maternally Derived Antibodies
Experimental Vaccines
Vectored Vaccines
Further Reading
References
Torque Teno Virus
Etiology
Epidemiology
Clinical Signs
Diagnosis
Further Reading
References
Nipah
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Necropsy Findings
Diagnosis
Treatment
Control
References
Tioman Virus
Reference
Porcine Retroviruses
Further Reading
References
Menangle
Etiology
Epidemiology
Clinical Signs
Necropsy Findings
Diagnosis
Differential Diagnosis
Treatment
Control
Further Reading
References
Japanese B Encephalitis (Japanese Encephalitis)
Etiology
Epidemiology
Pathogenesis
Clinical Signs
Necropsy Findings
Diagnosis
Control
Further Reading
Reston Virus
Reference
Bungowannah Virus
Porcine Parvovirus
Novel Porcine Parvoviruses
References
Multi-Organ Diseases Due to Protozoal Infection
Sarcocystosis (Sarcosporidiosis)
Etiology
Epidemiology
Occurrence
Source of Infection
Risk Factors
Climate
Species of Sarcocystis
Farm Dogs
Cats
Stocking Density
Economic Importance
Pathogenesis
Clinical Findings
Cattle
Sheep
Swine
Abortion and Perinatal Fatality
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Toxoplasmosis
Etiology
Epidemiology
Occurrence
Source of Infection
Cat Feces
Other Sources
Risk Factors
Pathogen Risk Factors
Environmental and Management Risk Factors
Experimental Studies
Sheep
Cattle
Other Ruminants
Pigs
Horses
Economic Importance
Zoonotic Implications
Pathogenesis
Pregnant Sheep and Goats
Clinical Findings
Sheep
Goats
Pigs
Cattle
Horses
Clinical Pathology
Abortion
Necropsy Findings
Abortion
Samples for Diagnostic Testing
Treatment
Control
Cat Control
Serologic Monitoring
Prophylaxis
Vaccination
Reduction of Zoonotic Risk From Food and Water
Further Reading
References
Theilerioses
References
East Coast Fever (ECF)
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Methods of Transmission
Risk Factors
Environmental Factors
Immune Mechanisms
Experimental Reproduction
Economic Importance
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Tropical Theileriosis (Mediterranean Coast Fever)
Etiology
Epidemiology
Occurrence and Methods of Transmission
Risk Factors and Immune Mechanisms
Economic Importance
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Multi-Organ Diseases Due to Trypanosome Infection
Nagana (Samore, African Trypanosomaisis, Tsetse Fly Disease)
Etiology
Epidemiology
Occurrence
Prevalence
Morbidity and Case Fatality
Methods of Transmission
Cyclical
Noncyclical
The Carrier State
Risk Factors
Host Factors
Environmental Factors
Pathogen Factors
Immune Mechanisms
Experimental Reproduction
Economic Importance
Zoonotic Implications
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Gross Pathology
Histology
Specimens for Pathology
Treatment
Control
Further Reading
References
Surra (Mal de Caderas, Murrina)
Etiology
Epidemiology
Occurrence
Morbidity and Case Fatality
Method of Transmission
Immune Mechanisms
Zoonotic Implications
Economic Importance
Biosecurity Concerns
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Multi-Organ Diseases Due to Fungal Infection
Protothecosis and Chlorellosis (Algal Bacteremia)
References
Coccidioidomycosis
Etiology
Epidemiology
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
References
Paracoccidioidomycosis (Paracoccidioides Infection)
References
Rhodotorulua spp. Infection
References
Histoplasmosis
References
Cryptococcosis (European Blastomycosis, Torulosis)
References
North American Blastomycosis
Further Reading
References
Multi-Organ Diseases Due to Metabolic Deficiency
Sodium and/or Chloride Deficiency
Magnesium Deficiency
Further Reading
References
Copper Deficiency
Etiology
Primary Copper Deficiency
Secondary Copper Deficiency
Epidemiology
Occurrence
Geographic Distribution
Primary Copper Deficiency
Secondary Copper Deficiency
Seasonal Occurrence
Risk Factors
Animal Factors
Age
Breed and Species Susceptibility
Fetal Liver Copper
Dietary Factors
Pasture Composition
Molybdenum and Sulfur
Copper in the Diet
Dietary Iron
Stored Feeds
Soil Characteristics
Copper Deficiency.
Molybdenum Excess.
Pathogenesis
Effects on Tissues
Changes in Gene Expression
Wool
Body Weight
Diarrhea
Anemia
Bone
Connective Tissue
Heart
Blood Vessels
Pancreas
Nervous Tissue
Reproductive Performance
Immune System
Development of Clinical Signs
Copper–Molybdenum– Sulfate Relationship
Copper Utilization
Hepatic Storage
Phases of Copper Deficiency
Clinical Findings
Cattle
Subclinical Hypocuprosis
General Syndrome
Primary Copper Deficiency
Secondary Copper Deficiency
Falling Disease
Peat Scours (“Teart”)
Unthriftiness (Pine) of Calves
Sheep
General Syndrome
Primary Copper Deficiency
Swayback and Enzootic Ataxia in Lambs and Goat Kids
Goats
Other Species
Deer
Pigs
Horses
Clinical Pathology
Herd Diagnosis.
Treatment Response Trial.
Copper Status of Herd or Flock.
Laboratory Diagnosis
Interpretation of Laboratory Results
Cattle and Sheep
Horses
Farmed Red Deer (Cervus Elaphus)
Necropsy Findings
Samples for Confirmation of Diagnosis
Treatment
Control
Dietary Requirements
Copper Toxicity
Copper Supplementation
Oral Dosing
Dietary Supplementation
Removal of Sulfates
Parenteral Injections of Copper
Slow-Release Treatments
Glass Bolus
Copper Oxide Needles
Sheep
Cattle
Farmed Red Deer
Genetic Selection
Summary and Guidelines
Further Reading
References
Riboflavin Deficiency (Hyporiboflavinosis)
Choline Deficiency (Hypocholinosis)
References
Multi-Organ Diseases Due to Toxicity
Snakebite
Etiology
Epidemiology
Risk Factors
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Bee and Wasp Stings (Hymenoptera)
Further Reading
Reference
Red Fire Ant Stings (Solenopsis invicta)
Further Reading
References
Moths
Sweating Sickness (Tick Toxicosis)
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
Reference
4-Aminopyridine Toxicosis
Further Reading
References
Cadmium Toxicosis
Further Reading
References
Chromium Toxicosis
Further Reading
References
Cobalt Toxicosis
Further Reading
References
Primary Copper Toxicosis
Etiology
Causes of Acute Oral Poisoning
Causes of Acute Injectable Poisoning
Causes of Chronic Oral Poisoning
Epidemiology
Occurrence
Sheep
Calves (Preruminant)
Cattle
Goats
Horses
Swine
Risk Factors
Animal Risk Factors
Species Susceptibility
Breed Susceptibility
Sheep.
Cattle.
Environmental Factors
Pathogenesis
Clinical Findings
Acute Intoxication
Chronic Intoxication
Clinical Pathology
Acute Ingestion
Chronic Ingestion
Necropsy Findings
Acute Intoxication
Chronic Intoxication
Samples for Confirmation of Diagnosis
Treatment
Control
Further Reading
References
Toxicosis From Dried Poultry Wastes
Toxicosis from Defoliants
Further Reading
Toxicosis from Fungicides
Fungistatic Agents
Grain Fumigants
Further Reading
Reference
Toxicosis from Herbicides
Bipyridyl Derivatives
Carbamates, Thiocarbamates, Dithiocarbamates
Dinitrophenol Compounds
Inorganic Herbicides
Sodium Chlorate
Organophosphorus Compounds
Phenoxy Acid Derivatives
Triazines/Triazoles
Ureas/Thioureas
Others
Triclopyr
Delrad
Further Reading
References
Hydrocarbon Toxicosis
Etiology
Epidemiology
Pathogenesis
Clinical Findings (Oil Ingestions)
Natural Cases
Experimental Cases
Clinical Pathology
Necropsy Findings
Treatment
Further Reading
Reference
Iron Toxicosis
Neonatal Pigs
Foals/Horses
Cattle
Further Reading
References
Toxicosis From Feed Additives
Bronopol
Carbadox
Pluronics
Tin Poisoning
Further Reading
References
Toxicosis from Miscellaneous Farm Chemicals
Formalin
Methyl Bromide
Polybrominated Biphenyls
Clinical Signs
Cattle
Pigs
Polybrominated Diphenyl Ethers
Polychlorinated Biphenyls
Sodium Fluorosilicate
Superphosphate Fertilizers
Further Reading
References
Toxicosis from Seed Dressings
Toxicosis from Miscellaneous Rodenticides
Bromethalin
Cholecalciferol (Vitamin D3)
Red Squill (Sea Onion)
Phosphides
Further Reading
References
Sulfur Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Transmission
Pathogenesis
Clinical Findings
Ruminants
Necropsy Findings
Treatment
Control
Further Reading
References
Vanadium Toxicosis
Further Reading
Reference
Toxicosis from Wood Preservatives
Chromated Copper Arsenate
Pentachlorophenol
Creosote (Coal Tar Creosote)
Further Reading
References
Zinc Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Farm Risk Factors
Transmission
Pathogenesis
Clinical Findings
Acute Poisoning
Cattle
Pigs
Chronic Poisoning
Dairy Cattle
Pigs
Horses
Clinical Pathology
Necropsy Findings
Samples for Confirmation of Diagnosis
Tissue Assay
Treatment
Control
Further Reading
References
Diterpenoid Alkaloid Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Ergot Alkaloid Toxicosis
Ergotism
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Peripheral Gangrene (Classical Ergotism)
Hyperthermia Form
Reproductive Form
Clinical Pathology
Necropsy Findings
Treatment
Control
Neotyphodium (Acremonium) spp. Toxicosis
Fescue Toxicosis
Clinical Findings
Fescue Summer Toxicosis (Summer Slump, Epidemic Hyperthermia)
Fescue Foot
Fat Necrosis (Lipomatosis)
Reproductive Abnormalities in Mares
Control
Further Reading
References
Fumonisin Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Environmental Risk Factors
Pathogenesis
Equine Toxicosis
Equine Leukoencephalomalacia (Moldy Corn Disease)
Clinical Findings
Clinical Pathology
Necropsy Findings
Swine Toxicosis
Porcine Pulmonary Edema
Clinical Signs
Necropsy Findings
Hepatosis/Hepatic Effects
Clinical Pathology
Necropsy Findings
Ruminant Toxicosis
Diagnosis
Treatment
Control
Further Reading
References
Glucosinolate Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Risk Factors
Human Risk Factors
Plant Factors
Pathogenesis
Clinical Findings
Goiter
Enteritis
Acute Pulmonary Emphysema and Interstitial Pneumonia
Polioencephalomalacia (Rape Blindness)
Other Unrelated Diseases
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Miscellaneous Mycotoxins
Cyclopiazonic Acid
Patulin
Sterigmatocystin
Further Reading
References
Mushroom Toxicosis
Amatoxins
Ramaria Flavo-Brunnescens
Scleroderma Citrinum
Cortinarius Speciocissimus
Inocybe and Clitocybe spp.
Further Reading
References
Phalaris spp. (Canary Grass) Toxicosis
Etiology
Epidemiology
Occurrence
Risk Factors
Animal Factors
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Treatment
Control
Further Reading
References
Toxicosis from Plant Phenols (Gossypol and Tannins)
Gossypol
Etiology
Epidemiology
Pathogenesis
Clinical Findings
Clinical Pathology
Necropsy Findings
Control
Tannins
Oak (Quercus spp.)
Yellow-Wood Tree (Terminalia Oblongata spp.)
Further Reading
References
Miscellaneous Plant Toxicosis
Aesculin
Alcohol (Complex Plant)
Aliphatic Acetogenin (Monoglyceride)
Amine Toxicity
Amino Acid Toxicity
Indospicine/Canavanine
Mimosine
Aristolochine
Crepenynic Acid
Cycad Glycosides
Grayanotoxins
Isoquinoline Alkaloids
Juniperine
Rhoeadine
Saponin Poisoning
Triterpene Saponins
Steroidal Saponins
Sesquiterpenes
Furanoid Sesquiterpenes (Furanosesquiterpenoid) Poisoning
Ipomeanol
Sesquiterpene Lactones
Vomiting Syndrome
Encephalomalacia Syndrome
Selenocompounds
Steroidal Alkaloids (Solanum spp.)
Vellein
Veratrine
Zigadine (Zigadenine)
Further Reading
References
Toxicosis From Brewer’s Residues
Trichothecene Toxicosis
Macrocyclic Trichothecenes
Stachybotryotoxicosis
Myrotheciotoxicosis
Nonmacrocyclic Trichothecenes
T-2 Toxin and HT-2 Toxin
Deoxynivalenol
Fusaritoxicosis Syndromes Without Specified Toxins
Further Reading
References
Triterpene Plant Toxicosis
Further Reading
References
Appendices
Appendix 1 Conversion Tables
Conversion Factors for Old and SI Units
Conversions
Temperature
Mass
Capacity
Length
Metric
US/Imperial
Pressure
Appendix 2 Reference Laboratory Values
Hematology
Hematology (International units, SI)
Serum constituents (U.S. units)
Serum constituents (International units, SI)
Appendix 3 Drug doses and intervals for horses and ruminants
Appendix 4 Drug doses and intervals for pigs
Index
A
B
C
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z
How to Use This Book
For Example
Guidelines for Selection and Submission of Necropsy Specimens for Confirmation of Diagnosis

Citation preview

VetBooks.ir

VetBooks.ir

VETERINARY MEDICINE A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs, and Goats

This page intentionally left blank

VetBooks.ir

VetBooks.ir

VETERINARY MEDICINE 11 EDITION

A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs, and Goats VOLUME ONE

PETER D. CONSTABLE KENNETH W. HINCHCLIFF STANLEY H. DONE WALTER GRÜNBERG

VetBooks.ir

3251 Riverport Lane St. Louis, Missouri 63043

VETERINARY MEDICINE: A TEXTBOOK OF THE DISEASES OF CATTLE, HORSES, SHEEP, PIGS, AND GOATS, ELEVENTH EDITION Copyright © 2017 Elsevier Ltd. All Rights Reserved. Previous editions copyrighted: 2007, 2000, 1999, 1994, 1983, 1979, 1974 First published 1960 No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions. This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

Notices Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary. Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility. With respect to any drug or pharmaceutical products identified, readers are advised to check the most current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the responsibility of practitioners, relying on their own experience and knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each individual patient, and to take all appropriate safety precautions. To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein. Main ISBN: 9780702052460 Volume 1 ISBN: 978-0-7020-7057-0

Content Strategist: Penny Rudolph Content Development Specialist: Laura Klein Content Development Manager: Jolynn Gower Publishing Services Manager: Hemamalini Rajendrababu Senior Project Manager: Kamatchi Madhavan Design Direction: Renee Duenow

Printed in China Last digit is the print number:  9  8  7  6  5  4  3  2  1

VetBooks.ir

Dr. Otto M. Radostits, August 31, 1934-December 15, 2006, Senior Author, Fifth to Seventh Editions; Lead Author, Eighth to Tenth Editions Otto Martin Radostits was a veterinary educator, clinician, and researcher who had a profound influence on students and practicing veterinarians throughout the world through his writings, not the least this text. Otto was closely involved with writing and editing the Fifth to Tenth Editions of Veterinary Medicine. Otto, the eldest son of Austrian immigrants, was raised on a small mixed farm in Alberta, Canada. His early farm experiences and those obtained from working with a local veterinarian while attending high school sparked an interest in pursuing a career in veterinary science and were the beginning of his lifelong passion for large-animal veterinary medicine. He was admitted to the Ontario Veterinary College in 1954, at that time the only English-speaking veterinary school in Canada. During his undergraduate years, his clinical interests and potential were recognized such that following graduation, he was invited to join the faculty as a member of the ambulatory clinic practice of the college—at that time a vigorous practice in a rural area. Otto spent the next 5 years teaching in this position, with the exception of a year spent at the veterinary school at Purdue University in West Lafayette, Indiana. The Western College of Veterinary Medicine in Saskatchewan, Canada, was established under the leadership of Professor D. L. T. Smith in the mid-1960s, and Otto was one of the founding faculty members. He established the ambulatory practice and helped design the college clinical buildings and finalize the curriculum. He remained a faculty member at the Western College of Veterinary Medicine until he retired in June 2002 and was awarded the title Emeritus Professor. Here he matured as a clinical teacher to influence students and veterinarians locally and internationally through his writings and presentations at veterinary meetings. Otto’s international recognition in large-animal veterinary medicine rests mainly on the strength of his writing and authorship of veterinary texts. These span the spectrum of large-animal veterinary medicine, from the clinical examination of the individual animal to the epidemiology, diagnosis, treatment, and control of livestock diseases, to herd health and preventive medicine. The most notable are his contributions to this textbook, which has been used by veterinary students and practicing veterinarians around the world for over 50 years and through 11 editions, for 6 of which Otto was a senior or lead author. Otto joined the original authors, Doug Blood and Jim Henderson, for the Fifth Edition of this text in 1979 and, in 1994, became the senior author for the Eighth, Ninth, and Tenth Editions. During his sojourn as senior author, the text continued its original design as a student textbook with many studentfriendly features. It also continued its importance as a reference book including the available information on all of the diseases of large animals, a truly formidable task. Otto did a large part of the work and would surely have been very proud of this new Eleventh Edition. In the writing of these and his other texts, Otto read the veterinary literature and was a firm believer in evidenced-based medicine. He insisted that all statements in these texts were supported by references in the literature, and he maintained the format of a very large bibliography at the end of each disease description. He believed that other veterinary educators should also be current with the veterinary

literature and had little regard for those who were not. He could be a forceful presence in discussions, but Otto was also one of the quickest to recognize new information that negated previous theories concerning a disease, and he was always responsive to reasoned argument. Otto taught that making a correct diagnosis was the crux to the solution of a disease problem, and he had a passion for the art and science of clinical examination. Many of his students affectionately remember his admonition, “We make more mistakes by not looking than by not knowing.” Otto’s insistence on the need for accurate diagnosis did not preclude this realization that what the practicing veterinarian needed as the final message from his books was the best current information on what to do to cure or prevent the disease in question. Otto has authored other texts. In the late 1990s he became concerned that the traditional skills of physical clinical examination were being supplanted by laboratory and instrumental analysis. As a consequence, he consulted with veterinary clinicians around the world and in 2000 was a senior author of the text Veterinary clinical examination and diagnosis. From his work on farms, Otto recognized that disease in farm animals commonly was a population concern, and he recognized the limitations of “fire brigade” medicine. He authored the first major text in herd health and preventive medicine with its first edition in 1985. Otto published many other works of significance to global veterinary medical education and presented more than 250 invited lectures and seminars in veterinary medicine in countries around the world. Dr. Radostits’s contributions have been recognized by many awards. For him, probably the most important were the award of Master Teacher from his university and, nationally, the Order of Canada.

Dr. Otto Radostits teaching at the Western College of Veterinary Medicine in Saskatchewan, Canada. (Image courtesy of Mrs. Ruth Radostits and family.)

v

Dr. Clive Collins Gay

VetBooks.ir

Dr. Clive Collins Gay, DVM (Guelph, 1960), MVSc (Guelph, 1962), MVSc (Ad Uendem Statum, Melbourne, 1970), FACVSc (1977), Diplomate of the American College of Veterinary Internal Medicine (honorary, 2008), and Doctor of Veterinary Science (Honoris Causa, Melbourne, 2008) has a distinguished career as an agricultural animal veterinarian, scientist, author, and educator spanning five decades. After graduating from Guelph in 1960, he was appointed as an assistant lecturer in the Department of Veterinary Medicine at the University of Glasgow from 1962 to 1964. In 1964, he was a George Aitken Pastoral Research Fellow (Sheep) and worked at the Veterinary Investigation Centre, Edinburgh University (Scotland); the Veterinary Investigation Centre, Ministry of Agriculture, Penrith (England); and the Nuffield Institute for Medical Research, Oxford University. In 1965, Dr. Gay was recruited by his mentor Professor Douglas Blood to the newly reestablished veterinary school at the University of Melbourne as a senior lecturer in agricultural animal medicine. Ken Hinchcliff was one of Dr. Gay’s students at the University of Melbourne. Dr. Gay was a genuinely gifted clinician, with an enthusiasm for veterinary science that inspired generations of undergraduate and postgraduate students and staff alike. His teaching attributes were recognized by various student accolades over the years, both in Australia and North America, and by the Washington State University (WSU) Faculty Award in 2000 from the Washington State Veterinary Medical Association (WSVMA). In 1979, Dr. Gay became a professor of food animal medicine at WSU, where he concentrated on agricultural animals, establishing the Field Disease Investigation Unit in 1982 and leading the unit until his retirement in 2005. The approach used by the Field Disease Investigation Unit was groundbreaking at the time it was implemented in that it applied a multidisciplinary approach including university and private veterinarians, animal scientists, extension agents, and producers to tackle economically important livestock diseases. Dr. Gay was also one of the earliest proponents of evidenced-based medicine. He served on several committees of the U.S. Department of Agriculture (USDA). In recognition of his extensive contribution in this area, he received the prestigious Calvin W. Schwabe Award for lifetime achievement in veterinary epidemiology and preventive medicine from the American Association of Veterinary Epidemiology and Preventive Medicine in 2007. Dr. Gay became Professor Emeritus at WSU in 2005. His extensive contribution to veterinary medicine was recognized with a Distinguished Achievement Award from the Washington State Veterinary Medical Association in 2006, and he was made an Honorary Diplomat of the American College of Veterinary Internal Medicine in 2008. Dr. Gay’s research activities covered the breadth of veterinary science in regard to both species and systems, including topics as diverse as colic in horses, cardiology in dogs, diarrhea in pigs, colostral immunity in calves, and trace-element deficiency in ruminants. He supervised 13 PhD students and 14 master’s degree students. This work resulted in more than 90 articles in journals, more than 100 proceedings and abstracts, and the delivery of more than 150 invited presentations to scientific groups, veterinary conferences, and agricultural groups. The latter reflected his commitment to “knowledge

vi

transfer” (extension work), which was a cornerstone of his approach to epidemiological studies and preventive medicine. Reflecting his international standing, Dr. Gay had been a Visiting Research Fellow in the following areas: the Department of Veterinary Microbiology, University of Guelph, in 1971; the Department of Veterinary Clinical Studies, University of Cambridge, in 1972; the Department of Veterinary Clinical Sciences, Massey University, in 1993; the Central Veterinary Laboratory, Ministry of Agriculture Fisheries and Food, Pirbright, in 1994; and the Department of Geospatial Science, RMIT University (Melbourne), in 2001. Over the years, Dr. Gay contributed actively to national and state veterinary associations, serving as a committee member of the Victorian Division of the Australian Veterinary Association (1968–1971); and editor of the Victorian Veterinary Proceedings (1968–1971); and an executive committee member of the Washington State Veterinary Medical Association (1999–2005), where he held the positions of vice president (2000), and president (2003–2004). Dr. Gay was a contributing author of Veterinary Medicine, edited by Blood, Henderson, and Radostits in 1979, 1983, and 1989, and an author and editor for the Eighth (1994), Ninth (2000), and Tenth (2007) Editions. His most important contributions to those editions included diseases of the newborn, infectious diseases of sheep and goats, prion diseases, practical antimicrobial therapy, and selected metabolic and protozoan diseases, emphasizing the important roles that environment, management, host factors, and pathogen virulence factors play in disease occurrence and severity. Dr. Gay was largely responsible for bringing the Tenth Edition to print when Dr. Radostits, the lead author and editor, became ill during the final stages of preparation of the text.

Dr. Clive Gay and Professor Doug Blood, Veterinary Clinical Centre, University of Melbourne, 1978. (Courtesy of D. Blood’s family.)

Professor Douglas Blood

VetBooks.ir

1920–2013 Professor Douglas Blood came to Australia in 1926 from East Ham, London. His family settled in Richmond, New South Wales, and toughed out the Great Depression. Through a scholarship, he attended Hurlstone Agricultural High School, where he enjoyed studying animals, especially cows and dogs. Following high school, Doug entered the Bachelor of Veterinary Science program at the University of Sydney. During World War II, he and a group of colleagues convinced the university to allow them to complete an accelerated course so that they could graduate in 1942 and then enlist in the armed services. Doug became a captain in a surveillance unit called Curtin’s Cowboys in the Northern Territory. He returned to teach at the University of Sydney Veterinary School for 12 years. Then, from 1957 to 1962, Doug taught large-animal medicine at the Ontario Veterinary College at Guelph. It was during this time that he taught and mentored Otto Radostits and Clive Gay, both of whom were subsequently to become authors, along with Doug, of this text. In 1962, Doug was appointed Professor of Veterinary Medicine and Founding Dean of Veterinary Science at the University of Melbourne. Doug passed on the deanship in 1968, but he continued to teach, retiring in 1985 after 23 years of service. During his time at the University of Melbourne, Doug recruited Clive Gay to a faculty (academic) position in the School of Veterinary Science and taught both Ken Hinchcliff and Peter Constable, both of whom followed him as authors of this text and deans of veterinary faculties—Hinchcliff at the University of Melbourne and Constable at the University of Illinois. In recognition of his service to veterinary science, Doug was the recipient of many awards and honorary degrees, including the Schofield Medal from the University of Guelph, the Gilruth Prize for Meritorious Service to Veterinary Science from the Australian Veterinary Association, and an Order of the British Empire. He was involved in the formation of the Australian and New Zealand College of Veterinary Scientists. He also served as a committee member of the Victorian Division of the Australian Veterinary Association and as a board member of the Veterinary Surgeons Registration Board of Victoria. In the early years, Doug Blood revolutionized the teaching of clinical veterinary medicine. For those of us privileged to have been taught by him at this time, he was a superlative teacher. Doug was one of the first teachers in clinical veterinary medicine to recognize that pathophysiology was the basis for teaching the disease processes in large animals. He also concentrated on the principles of pathophysiology in his explanations of disease syndromes and in teaching clinical examination and diagnosis. This was an approach that he developed from the teaching of his mentor, Oxford veterinary scientist H. B. Parry, to whom the first edition of this text was dedicated. This approach to clinical teaching was in marked contrast to the rote learning that was common in many of the disciplines taught at that time and in stark contrast to the teaching method in clinical examination and diagnosis, which primarily relied on pattern recognition. Doug Blood also taught that the method of clinical examination should be system based, that it should be conducted in a systematic manner, and that it should be conducted using all available senses and techniques. He further taught that the intellectual diagnostic rule-out process should also incorporate a consideration of the presenting epidemiology of the disease problem, an examination of the environment, and an estimation of the probability of disease occurrence, summarized with his often repeated adage “common diseases occur commonly.” Although these approaches might seem obvious to recent graduates, in the 1950s and early 1960s, they were revolutionary. In fact, they set the foundation for current teaching principles in largeanimal clinical veterinary medicine. Students of that older vintage recall with great appreciation the understanding of clinical veterinary medicine imparted by Doug Blood and his particular contribution to

Professors Ken Hinchcliff, Peter Constable, and Doug Blood (Werribee, Australia, 2008). (Source: Hinchcliff K.)

their education. Throughout subsequent years in his teaching career, Doug had the ability to inspire students and is viewed with respect, admiration, and even veneration by the generations of students he has taught. The First Edition of this text was published in 1960 and authored by D. C. Blood and J. A. Henderson. It was entitled Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs and Goats and was based on Doug Blood’s and Jim Henderson’s lectures and Doug’s teaching and philosophical approach. At that time, there were few textbooks in the disciplines of veterinary science and up-to-date texts published in English that were primarily concerned with clinical veterinary medicine and diseases in agricultural animal species were not available. The original text was divided into two major sections. One section, “General Medicine,” covered system dysfunction, and the other, “Special Medicine,” covered the specific diseases of the largeanimal species. This format was followed until the Eleventh Edition. The Second Edition was published in 1963 and had an additional two chapters covering parasitic diseases. Subsequently, new editions have been published approximately every 5 years, with major or minor changes in format in most editions, such as the addition of chapters dealing with new subjects or the addition of material in specific subheadings to highlight, for example, the epidemiology or zoonotic implications of disease. However, always, with each edition, there was an extensive revision of disease descriptions based on current literature. Professor Henderson’s involvement with the text ceased with the Fifth Edition, and that edition recruited Professor O. M. Radostits as senior author and others as contributing authors. Blood coauthored nine editions over a span of 45 years, with coauthors including Radostits, Gay, Hinchcliff, and Constable. In the preface to the First Edition, it was stated that the book was directed primarily to students of veterinary medicine, although it was expected that the book would be of value to practicing veterinarians and field workers. The latter expectation has certainly proved true, and the book has come to be extensively used as a reference by veterinarians in large- and mixed-animal practice around the Englishspeaking world. Editions of the text have also been translated into French, Italian, Spanish, Portuguese, Japanese, Chinese, and Russian. In addition to his passion for the method and accuracy of diagnosis of disease in individual animals and herds, Doug Blood also had a passion for preventive medicine and was a firm proponent of the thesis that subclinical disease is economically more important than clinical disease in agricultural animal populations. With other vii

viii

Professor Douglas Blood

VetBooks.ir

colleagues at the University of Melbourne, he developed health programs for dairy cattle, beef cattle, and sheep and conducted practical trials of these programs in private herds and flocks. These programs were based on a whole-farm approach and centered on the concept that performance targets could be tracked through computer-based productivity monitoring to detect deviation from target performance. Doug Blood was a very early proponent of the use of computers to manage and analyze data in clinical diagnosis and herd health man-

agement. These herd health programs have been successfully commercially adopted in several countries. Doug had a formidable intellect combined with an inexhaustible work ethic. He was a generous family man who had a zest for life and dry wit and who was so proud of his family and their achievements. He loved his morning runs/walks with his beloved Border Collies, music, and literature, and Doug had a passion for baking bread, brewing beer, photographing birds, and wearing bow ties.

VetBooks.ir

Contributors The authors’ names, degrees, and contact emails are below.

Authors Peter D. Constable BVSc(Hons), MS, PhD, Docteur Honoris

Contributing Authors

Causa (Université de Liège), Dipl.ACVIM, Dipl.ACVN (Honorary), AssocMember.ECBHM; ([email protected]) Professor and Dean, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA

D.D. (Doug) Colwell BSc, MSc, PhD; (Doug.Colwell@AGR.

Kenneth W. Hinchcliff BVSc(Hons), MS, PhD, Dipl.ACVIM;

Sara Connelly, DVM, MS, Dipl.ACVCP; ([email protected])

[email protected]; President and Chief Executive Officer, Trinity College, University of Melbourne, Royal Parade, Parkville, Victoria, 3052, Australia; past Dean Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Werribee, Victoria, Australia

Stanley H. Done BA, BVetMed, DVetMed, Dipl.ECPHM,

FRCVS, FRCPath; ([email protected]) Animal Health and Veterinary Laboratories Agency (AHVLA), Thirsk, United Kingdom. Contact [email protected]

Walter Grünberg DrMedVet, MS, PhD, Dipl.ECBHM, Dipl. ECAR, AssocDipl.ACVIM; ([email protected]) Farm Animal Internal Medicine Specialist, University of Veterinary Medicine Hannover, Foundation Clinic for Cattle, Hannover, Germany

GC.CA) Livestock Parasitology/Parasitologie du betail, Agriculture and Agri-Food Canada/Agriculture et Agroalimentaire Canada, Lethbridge Research Centre, Alberta, Canada

Clinical Assistant Professor, Department of Pathobiology and Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Illinois, Illinois, USA

Levent Dirikolu DVM, MVSc, PhD; ([email protected]) Profes-

sor and Director of the Equine Medication Surveillance Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA

Robin Gasser Tierarzt, DVM, PhD, DVSc; (robinbg@unimelb.

edu.au) Professor in Veterinary Parasitology, Veterinary Preclinical Centre, Faculty of Veterinary Science, University of Melbourne, Parkville, Victoria, Australia

Lynn Hovda RPh, DVM, MS, Dipl.ACVIM; (lhovda@safetycall.

com) Adjunct Professor, University of Minnesota College of Veterinary Medicine, Director of Veterinary Services, SafetyCall International and Pet Poison Helpline, Bloomington, Minnesota, USA

Basil Ikede DVM, PhD, Diagn Path, FCVSN; ([email protected]) University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada

John Larsen BVSc, PhD, Grad Dip Bus Admin; (j.larsen@unimelb.

edu.au) Associate Professor of Ruminant Production Medicine and Director, The Mackinnon Project, Veterinary Clinical Centre, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 250 Princes Highway, Werribee, Victoria, Australia

William Witola BVetMed, MSc, PhD; ([email protected])

Assistant Professor of Parasitology, Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA

Amelia R. Woolums DVM, MVSc, PhD, Dipl.ACVIM, Dipl.

ACVM; ([email protected]) Professor, Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi, USA

ix

VetBooks.ir

Preface to the Eleventh Edition We are delighted to present the Eleventh Edition of Veterinary Medicine, 56 years since the first “Blood and Henderson” Veterinary Medicine was published in 1960 and 9 years since the Tenth Edition was published in 2007. Veterinary Medicine focuses on diseases of ruminants, horses, and swine, and it is the most extensively cited textbook in veterinary medicine, with a recent total of 4,267 citations (Google Scholar, May 2016). Because the demand for this book remains strong, we assume that we have developed a philosophy, format, and price that are attractive and meet the demands of undergraduate veterinary students and graduate veterinarians working in the field of largeanimal medicine. Substantial changes were made to the format of the book for the Eleventh Edition to keep current with the continuing expansion of knowledge about the diseases of large animals. The book has been extensively revised and reorganized based on the major organ system affected. The organ systems approach reflects the profound impact that Dr. D. C. Blood had on the practice of large-animal medicine worldwide (see Foreword); he emphasized that the clinical examination procedure should be a systems-based method. We have extended the systems approach implemented in the First Edition through the assignment of diseases to the primary organ system affected or the most obvious clinical sign referable to an organ system. As a result, the Eleventh Edition contains 21 chapters, compared with 36 chapters in the Tenth Edition. Thirteen chapters deal with specific organ systems, including the alimentary tract of ruminants and nonruminants; the liver and pancreas; and the cardiovascular, hemolymphatic/ immune, respiratory, urinary, nervous, musculoskeletal, and reproductive systems; in addition to metabolic/endocrine abnormalities, diseases of the mammary gland, and, finally, diseases of the skin, eye, and ear. Each of these chapters is organized in the following manner: general diseases; infectious diseases, listed in order of cause (bacterial, viral, prion, protozoal, fungal, metazoan) and species affected (all large animals, ruminants, horses, pigs); metabolic diseases; nutritional diseases; toxicologic diseases and environmental agents; neoplastic diseases; congenital and inherited diseases; and, finally, diseases of unknown etiology. The remaining eight chapters deal with specific medicine topics, as follows: clinical examination and making a diagnosis; examination of the population; biosecurity and infection control; general systemic states; disturbances of free water, electrolytes, acid-base balance, and oncotic pressure; practical antimicrobial therapeutics; perinatal diseases, and systemic and multi-organ diseases. A comprehensive index permits the reader to easily access relevant information in different chapters of the book. We have attempted to ensure that the book continues to have an international scope by including clinically important diseases occurring in large animals worldwide. The book notes the eradication of Rinderpest in 2011 and includes new or extensively revised sections on a variety of topics, such as biosecurity and infection control; the Schmallenberg and bluetongue viral epidemics of ruminants in Europe; Wesselsbron disease in cattle and hypokalemia in adult cattle; equine multinodular pulmonary fibrosis; Hendra virus infection; multisystemic, eosinophilic, epitheliotropic disease of horses; hypoglycin A intoxication and equine metabolic syndrome; porcine reproductive and respiratory syndrome; porcine epidemic diarrhea and circovirus, and malignant catarrh in pigs; Torque teno, Menangle, and Japanese B viruses in pigs; and numerous recently identified congenital and inherited disorders of large animals. Reflecting the international scope of the book, the four authors and nine coauthors were educated or have practiced veterinary medicine in 12 countries covering five continents, including Australia, Austria, Canada, Germany, Japan, the Netherlands, Nigeria, Turkey, Switzerland, the United Kingdom, the United States, and Zambia. x

We continue to emphasize the epidemiology and pathophysiology of each disease, which are important in understanding the rationale for the diagnosis, treatment, and control. This means that we strive to maintain an optimum balance between published research and what field veterinarians find useful in their daily work. To make it easier for the reader to find particular pieces of information, long passages of prose have been divided into smaller sections using headings and subheadings. Key words, terms, and phrases have been emboldened for emphasis and to make it easier for the reader to identify important points. We also continue to include the zoonotic and bioterrorism implications of many diseases and how the largeanimal veterinarian is becoming more involved in the control of diseases transmissible to humans. The use of individual diagnostic tests, described under Clinical Pathology for each disease, continues to be a challenge for all of us, especially with the increased availability of genomic or genetic testing and point-of-care testing. We have continued to concentrate on those tests that are accepted through common use, to discuss their limitations if they are known, and to provide a reference to newer tests that have future promise in diagnosis. A common limitation of publications describing new diagnostic tests is the absence of, or inadequate, information on the characteristics (sensitivity, specificity, accuracy) of the test in the population of animals in which it will be used. Consistent with our deep commitment to practicing evidencedbased veterinary medicine, relevant references from 2006 onward have been cited, and important review and scientific papers, including Internet sites, are identified as Further Reading. We refer readers to previous editions of the book for references to earlier works. When permitted by the quality and number of peer-reviewed publications, we have applied the Grading of Recommendations Assessment, Development and Evaluation (GRADE) process (see Foreword) to provide a summary of treatment and control recommendations in a box at the end of the section. This process distills information down to one of four recommendations that reflect “a judgment that most well-informed people would make”: R1, “do it”; R2, “probably do it”; R3, “probably don’t do it”; and R4, “don’t do it.” We believe that the GRADE approach will prove helpful to large-animal veterinarians, and we look forward to expanding this approach in future editions of this book. Constraining the size of the book has been a constant preoccupation and a difficult task with the ever-increasing volume of published information and the constantly growing list of diseases. Our intention has always been to provide information on all recorded diseases. Despite of reductions in reference lists and extensive editing to minimize repetition, the book is still large, necessitating a move to two volumes. More than 150 new figures have been added to the book to assist in presenting information. We continue to subscribe to the practice and philosophy of earlier editions of this book in having a small number of authors contribute the majority of the text, with contributions from content specialists for particular topics. We believe that analysis and review of the relevant literature by a small number of authors with a broad knowledge and global perspective of large animal medicine assures a consistency of approach to each topic. Our authors are based in the United States, Australia, Europe, the United Kingdom, and Canada and have extensive experience in international veterinary medicine. Dr. Peter Constable, Dean of the College of Veterinary Medicine, University of Illinois, USA, has assumed the responsibilities of senior author. He revised a number of sections related to specific ruminant diseases, in addition to major sections of the chapters on general systemic states and diseases of the ruminant alimentary tract, cardiovascular system, urinary system, musculoskeletal system, nervous system, and mammary gland. Dr. Constable also revised the chapters

Preface to the Eleventh Edition

VetBooks.ir

on examination of the population and disturbances of free water, electrolytes, and acid-base balance. Dr. Kenneth Hinchcliff, CEO of Trinity College, University of Melbourne, and former Dean of the Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia, revised all the equine diseases and major sections of the chapters on diseases of the respiratory system, nonruminant alimentary tract, hemolymphatic and immune systems, endocrine abnormalities, and diseases of the neonate. Dr. Hinchcliff also revised the chapter on clinical examination and making a diagnosis and the Foreword of the book. Dr. Hinchcliff acknowledges the support of St. John’s College, Cambridge, in appointing him as Overseas Visiting Scholar in 2013 during preparation of parts of this text. Drs. Constable and Hinchcliff are responsible for the revised format of the book. Dr. Stanley Done, recently retired from the Animal Health and Veterinary Laboratories Agency, Thirsk, United Kingdom, joined our book as a coauthor and revised all the sections on diseases of pigs. This was a major task given the very large literature base on infectious diseases of pigs on a worldwide basis. Dr. Walter Grünberg, Farm Animal Internal Medicine Specialist, Tieräerztliche Hochschule, University of Veterinary Medicine, Hannover, Germany, is also a new coauthor. He revised a number of sections related to specific ruminant diseases and extensive sections of the chapters on diseases of the liver and pancreas and the skin, eye, conjunctiva, and ear. The legacies of Drs. D. C. Blood, C. C. Gay, J. A. Henderson, and O. M. Radostits continue in this edition of Veterinary Medicine. Dr. Doug Colwell, Principal Research Scientist at Agriculture and Agri-Food Canada, once again revised the sections on diseases caused by arthropod ectoparasites. Dr. Sara Connelly, Clinical Assistant Professor of Clinical Pathology, College of Veterinary Medicine, University of Illinois, USA, revised the appendices dealing with conversion tables and reference laboratory values. Dr. Levent Dirikolu, Professor of Pharmacology, School of Veterinary Medicine, Louisiana

xi

State University, USA joined our book as a contributor by revising the chapter on practical antimicrobial therapeutics. Dr. Robin Gasser, Professor of Veterinary Parasitology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia, is a new contributor who revised the coverage of protozoal diseases. Dr. Lynn Hovda, Director of Veterinary Services, PLLC and Pet Poison Helpline, Minnesota, USA also joined our book by revising the sections related to diseases caused by toxins in plants, fungi, cyanophytes, clavibacteria, and venoms in ticks and vertebrate animals. Dr. Basil O. Ikede, recently retired from the Atlantic Veterinary College in Prince Edward Island, Canada, once again revised the sections on the major exotic viral and protozoan diseases. Dr. John Larsen, Director of the Mackinnon Project, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Australia, is a new contributing author and revised many chapters related to diseases of sheep and goats. Dr. William Witola, Assistant Professor of Parasitology, College of Veterinary Medicine, University of Illinois, USA, is also a new contributor to the book, revising chapters related to nematode, trematode, and tapeworm parasitic infection. Dr. Amelia Woolums, Professor of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, USA, joined the Eleventh Edition by authoring a new chapter on biosecurity. We believe that we have completed another authoritative and comprehensive review of the peer-reviewed literature of large-animal medicine, at a standard at least equal to that of the previous 10 editions. We hope that the Eleventh Edition of Veterinary Medicine provides the information necessary to meet the needs of veterinary students and large animal clinicians for the next 5 to 8 years. P. D. Constable K. W. Hinchcliff S. H. Done W. Grünberg

VetBooks.ir

Introduction The first edition of this book established its role as a textbook of the diseases of traditional farm animals in the Western world, those being cattle, horses, sheep, pigs, and goats. The primary objective of this book was to offer the veterinary student and the practitioner the knowledge and information necessary to provide animal health management for farm animals. Although this intent has not changed, the context of veterinary medicine and large-animal practice has changed markedly in the 56 years since publication of the first edition.

VETERINARY MEDICINE IN THE ANTHROPOCENE Anthropocene is the proposed name for a new, and the current, geological epoch following on from the Holocene and demarcated as the time when human activities began to have a substantial global effect on the Earth’s systems.1,2 Although not universally accepted, the proposal recognizes that human activity has become the primary determinant of Earth’s biophysical conditions, influencing global systems and having profound effects on local and regional environments. The Anthropocene is also associated with marked political and economic changes, including regional instability and reductions in political or economic barriers to trade. All of these factors have influenced, and will continue to influence, veterinary practice and the management of the health, well-being, and productivity of animals used for production of fiber and human food.3 The concept of the Anthropocene allows veterinarians to consider how the veterinary profession will adapt to our changing environment and associated social, political, environmental, and economic challenges to animal and human health. The challenges include, but are not limited to the following:4 • A changing climate with flow-on effects on the geographic distribution of diseases, emergence or reemergence of infectious and noninfectious diseases, and extension of diseases into species not historically affected • Altered farming patterns, and hence use of animals, as climatic changes force farmers to abandon decades or centuries old land and animal management practices • Increasing internationalization of trade and freedom of movement of people, animals, and potential fomite, with important implications for the biosecurity of countries, regions, and industries • Political instability, with subsequent loss of animal health monitoring and disease control • Economic pressures to produce more and safer food with no increase in water or land use • Societal expectations for increased animal welfare and the associated mandated changes in farm animal management practices, for example, housing of dairy cattle or mulesing of lambs As noted by the “Safeguarding Human Health in the Anthropocene Epoch: Report of the Rockefeller Foundation-Lancet Commission on Planetary Health,”2 the scale of human impact on the planet is immense and includes the following changes: • About a third of the ice-free and desert-free land surface of the planet has been converted to cropland or pasture. • Annually, roughly half of all accessible freshwater is appropriated for human use. • More than 2.3 million km2 of primary forest has been cleared since 2000. • More than 60% of the world’s rivers are dammed, affecting in excess of 0.5 million km of river. • Extinction rate of species is more than 100 times that observed in the fossil record, and many remaining species are decreasing in number. xii

• Concentrations of major greenhouse gases—carbon dioxide, methane, and nitrous oxide—are at their highest levels for at least the past 800,000 years. • The global temperature continues to rise above long-term historic levels, driving changes in climate and weather patterns. These changes have a profound impact on human and animal health, as evidenced by altered geographic distributions of diseases, emergence of new diseases, and reemergence of previously controlled or repressed diseases. Anthropogenic changes in the environment influence animal health by affecting the productivity of agricultural and animal production systems and increasing the likelihood of spread of diseases from animals to humans. One-half of the global emerging infectious disease events of zoonotic origin between 1940 and 2005 are estimated to be the result of changes in land use, agricultural practices, and food production practices.5 There is evidence of an increased risk of zoonotic disease transmission in disturbed and degraded habitats, as exemplified by the emergence of two diseases caused by henipaviruses in animals and humans. Both Nipah and Hendra henipaviruses “spill over” from bats to pigs and horses, respectively, and subsequently infect humans. In both instances, disease is associated with altered habitats, including land clearing that creates a pathway for the repeated transmission of virus from fruit bat reservoirs, emphasizing the role of intact ecosystems and the suitability of climatic conditions in regulating the transmission of diseases.6-8 Change in weather systems and climate can profoundly influence the distribution of vectors for important pathogens. Climate is a major factor in determining the geographic and temporal distribution of arthropods, characteristics of arthropod life cycles, dispersal patterns of associated arboviruses, the evolution of arboviruses, and the efficiency with which arboviruses are transmitted from arthropods to vertebrate hosts.9 For example, emergence of bluetongue virus infection and disease in Europe has been attributed to climate change– induced alterations in distribution of hematophagous midges that transmit the virus, although this concept is disputed, and the importance of other anthropogenic, vector, or virus factors in the spread of the disease is unclear.10-13 There is concern that changes in land-use patterns and climate might provide for the spread of Rift Valley fever into new locations, including more frequent or deeper incursions into the Arabian Peninsula.14 The emergence of Schmallenberg virus, an orthobunyavirus, as a cause of disease in ruminants in Germany and the Netherlands in late 2011, and its subsequent rapid spread across Europe, highlights the potential for emergence of new diseases. The virus is apparently spread by culicoides midges, but its origin remains unknown. Its roughly concurrent occurrence with new strains of bluetongue virus (e.g., BTV-6) in the same region might be more than a coincidence.15 International transport of animals and animal products has the proven potential to introduce disease into areas in which it was not present. Introduction of African horse sickness into regions with populations of susceptible horses and competent midge (Culicoides spp.) vectors can result in spread of the disease, as occurred in the Iberian Peninsula in 1987.16-18 Equine influenza virus was inadvertently introduced into Australia in August 2007 by importation of infected horses from Japan, with spread occurring because of apparently inadequate quarantine procedures.19 Incursion of the virus, which was subsequently eliminated from the continent, had an adverse economic impact.20 Similarly, political instability and conflict, which might or might not be associated with climate change but in either case is clearly human-related, results in altered patterns of human movement, loss of control and eradication programs, and absence of surveillance, with

Introduction

VetBooks.ir

resultant resurgence of previously repressed diseases. Examples include the incursion of rinderpest into Turkey from Iraq in the aftermath of the first Gulf War in the early 1990s, and spread of bovine contagious pleuropneumonia in Angola and the Horn of Africa as a result of civil wars.21 It is not just in infectious diseases that we will see changes in the Anthropocene. Increasing global temperatures and altered rainfall conditions will increase the potential for heat- and drought-associated illnesses in farm animals and for disruption to social systems by altered rainfall patterns causing flooding, droughts, and an increase in extreme weather events.4 For instance, heat stress profoundly influences milk production, weight gain, and fertility of cattle, with these effects extending beyond the period of actual exposure to heat.4 Heat stress is a concern for dairy, feedlot, and range cattle in temperate and tropical regions of much of the world and has led to the introduction of management systems to accommodate the changes in weather.3,22,23 Increases in mean global temperature will increase the number of days annually, in some instances by over 100%, on which cattle are exposed to conditions that will cause heat stress.4 Veterinary input to animal productions systems will need to reflect these complex and changing climatic, political, social, and economic environments that represent the nature of contemporary livestock production of fiber and food for human consumption.

CONTEMPORARY LIVESTOCK PRODUCTION Although traditional farms incorporating multiple livestock systems still exist, much more important economically and in terms of the numbers of animals involved are farms that concentrate on one or two livestock systems or species. For example, witness the almost absence in developed countries of farms on which pigs are run in extensive systems on pastures or fields shared with cattle, horses, or sheep. Much more common, and economically important, are the large piggeries that house hundreds to thousands of pigs, often with farms focusing on breeding pigs that are then transferred to other farms or facilities for finishing. Or consider the ascension of feedlots in which sometimes massive numbers of cattle are aggregated from the many individual farms on which they were bred, or the change in dairies from small (50–100 cows) farms to large operations with thousands of cows. The disease issues confronting managers and their veterinary advisors in these facilities are much different from those encountered by a veterinarian responding to a call for a sick cow in a small dairy herd on a family-run farm that also produces sheep, pigs, and poultry. This edition of Veterinary Medicine reflects these changing circumstances, which are discussed in more detail in this Introduction. Another important change confronting veterinarians is the increasing value of some individual animals, particularly horses. The manner in which this book deals with medicine of horses is discussed in detail during the planning of each edition. Increasing value of individual horses and desire by owners to protect the health or performance of these animals have driven the veterinary profession to develop sophisticated, and expensive, diagnostic and therapeutic modalities and interventions directed toward care of individuals. However, this is offset by the recognition that preservation of the health of bands of horses on studs or in stables, or in whole populations in a country, is economically important and based on a thorough understanding of epidemiology and biosecurity—just as for food- and fiber-producing animals. For this latter reason, equine medicine in this book is dealt with as much from a population perspective as it is from the perspective of diagnosis and treatment of an individual horse. It is stating the obvious that veterinary medicine has advanced in ways that were unimaginable 56 years ago. For example, our understanding of the genetic, and increasingly the genomic, basis of production and susceptibility to disease is something that is now included in discussions of almost every disease, and not just those diseases with a clear monogenetic basis. Associated with this is the incalculable value of use of diagnostic tests based on detection of all or part of the

xiii

genome of a pathogen. Polymerase chain reaction tests now allow detection of infinitesimally small quantities of DNA or RNA with great rapidity and absolute specificity, often permitting detection of the presence of pathogen genetic material on the same working day as the samples were collected. Additionally, analysis of part (such as single-nucleotide polymorphisms, or SNIPs) or all of the genome of an animal or pathogen provides information critical to understanding the pathogenesis, pathogenicity, or epidemiology of the organism. The utility of this type of information is evident in discussion of many of the diseases—from detection of mutations in the genome of animals that cause them to display particular diseases, for example, bovine leucocyte adhesion deficiency, or, conversely, decrease their susceptibility to infectious diseases, such as occurs with scrapie in some breeds of sheep; to understanding of the pathogenicity of microbes, such as equine herpesvirus 1 neuropathic and less neuropathic strains, or their epidemiology, such as in strain typing of equine influenza H3N8 viruses.

VETERINARY CLINICAL EPIDEMIOLOGY Important to our understanding of the basis of disease has been the emergence of clinical epidemiology as a means of interrogating the patterns of disease and disease spread and identifying risk factors for development of disease. Understanding patterns of disease spread is fundamental to developing and implementing sensible and effective biosecurity and control measures. Similarly, knowing the risk factors for development of disease and quantifying the relative importance of each (“relative risk” or “odds ratio,” depending on the context) are key to determining which of these factors can be modified to reduce risk of the disease and whether it is economical to do so. The importance of use of applied and analytical epidemiology in large-animal practice and veterinary medicine is clear. The tools of epidemiology are now readily available to allow the veterinarian to identify and quantify the risk factors associated with the disease, to provide a more accurate prognosis, to accurately assess treatment responses and not depend on clinical impressions, to scientifically evaluate control procedures, and to conduct response trials. There is a large and challenging opportunity for veterinarians to become involved in clinical research in the field where the problems are occurring. It will require that they become knowledgeable about the use of computerized databases. These now provide an unlimited opportunity to capture and analyze data and generate useful information, which heretofore was not considered possible. The technique of decision analysis is also a powerful tool for the veterinarian who is faced with making major decisions about treatment and control procedures.

VETERINARY SCIENTIFIC LITERATURE AND HOW TO USE IT Perhaps the single greatest advance in veterinary medicine has been the collective increase in knowledge. The large increase in knowledge of animal diseases and animal health, including information about efficacy of diagnostic and therapeutic techniques and interventions, coupled with the ease of access of this information through online databases and web-based search engines, presents challenges in assessing the quality of information and in collating the information into a useable form. Development of formal methods for assessing information and providing a recommendation have led to the term evidence-based veterinary medicine.24-26 Evidence-based veterinary medicine is defined as the use of best relevant evidence in conjunction with clinical expertise to make the best possible decision about a veterinary patient, taking into account the circumstances of each patient and the circumstances and values of the owner/carer.27 The questions related to use of an evidence-based approach can be summarized as follows: • Why do we need evidence of effectiveness of our clinical actions (assessment of clinical signs, diagnostic tests, interventions, prognostication)?

xiv

Introduction

VetBooks.ir

• What are the levels of evidence, and how good are they? • How does one translate evidence into a recommendation or decision? • What factors contribute to the weighting of evidence? • Does weak evidence allow us to make a strong recommendation? • Does strong evidence not always allow us to make a strong recommendation? • How do I use this in practice? There are five steps to evidence-based veterinary medicine:27 1. Ask the pertinent question and thereby define what it is that needs to be known to allow for the most appropriate action. 2. Acquire the evidence, usually by a review of the available literature or, less commonly, performing a new research study. 3. Appraise the quality of evidence and its external validity (evidentiary value for the question being asked). 4. Apply the evidence to practice, where appropriate. See comments that follow about the GRADE process. 5. Audit—Assess whether the application of the new evidence has affected the outcome of interest. Quality of Evidence The confidence we have in our evidence-based approach to veterinary practice depends on our assessment of the quality of the evidence available to us.28 Not all evidence is of equal merit or utility, and although there are slight differences in the ratings of quality of evidence from different sources, an approximate hierarchy of evidence from lowest to highest in terms of value of evidence for practical use is as follows: • Expert opinion/editorials/nonstructured consensus statements or opinion pieces • Case reports and case series • In vitro studies with an appropriate control group • Animal models of the disease of interest (induced disease in species other than the species in which the disease occurs naturally, e.g., mouse model of a disease in horses) • Case-control or cross-sectional studies • Nonrandomized trials, cohort studies, or models of induced disease in the species of interest (target species, e.g., induction of viral diarrhea in calves) • Randomized controlled trials under field conditions • Systematic review of randomized controlled trials • Systematic review, including meta-analysis The higher the quality of evidence, the greater is our confidence in making decisions based on this evidence. The highest-quality evidence is provided by systematic reviews, which might include a metaanalysis. Systematic reviews differ from narrative reviews, which have much lower evidentiary value, in that systematic reviews are approached in a manner and with methodology designed to ensure the validity of the conclusions.29 Systematic reviews should be based on a clearly defined question and prespecified criteria for inclusion and evaluation of the literature, among other factors. Criteria and methodology for performing systematic reviews are available.30-32 Assessment of the quality of evidence provided by scientific articles is dependent on the authors of the article reporting exactly what they did and how they did it. It is clear from studies in human medicine and small animals that poorer reporting of methodology in articles is associated with a greater proportion of positive outcomes, leading one to suspect that reports that are less well documented are more likely to provide unreliable evidence of efficacy.33 There are increasing numbers of guidelines that provide advice for authors on how to adequately report on trials, and these guidelines are also useful as checklists for readers of articles. Available guidelines are CONSORT, REFLECT, STARD, STROBE, and others, which are available through the EQUATOR website (http://www.equator-network. org/reporting-guidelines/).34

From Evidence to Recommendation The approach of using evidence to guide clinical decision making has been formalized in the last two decades in human medicine and is gaining traction in veterinary practice. As veterinary clinicians, we have ethical and legal obligations to use methods and practices that are most likely to provide the “best” outcomes for the animals we treat and their owners. A traditional approach to deciding on the “best” treatments, diagnostic tests or methods, and preventative measures has been to identify the highest-quality evidence of effectiveness and to adopt the approach with the strongest evidence of efficacy. The Cochrane Collaboration and the Cochrane Reviews exemplify and lead this approach in human medicine (http://www.cochrane.org/ cochrane-reviews). This “evidence-based” approach has the implicit assumption that one should rely on the highest-quality evidence and that high-quality evidence of efficacy necessarily leads to the adoption of that treatment, diagnostic test, or prophylaxis. However, this approach falls short when formulating recommendations for use in clinical practice. What practitioners need is recommendations that are based on the available evidence but that also take into account the other factors that must be considered when advising an owner or trainer on the “best” approach to dealing with their animal’s (and their) problem. This methodology has been developed in human medicine as the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) process.35 GRADE fundamentally works by providing a framework to determine a final recommendation on an intervention through use of the following: 1. The quality of evidence (Cochrane and similar evaluations of quality of evidence stop here), 2. Seriousness of the outcome, 3. Magnitude of the treatment effect, 4. Precision of the treatment effect, 5. Risk of the target event (how frequent), 6. Risk of adverse events associated with the intervention, 7. Cost of the intervention, and 8. The values and preferences of the end users (patients). All of these criteria have at least some applicability in veterinary medicine. Briefly, judgments about the quality of recommendations require consideration of the following factors: • The quality of evidence on which the recommendation is based. The quality of evidence is assessed on the type of study (with systematic reviews being designated a priori as the highest level of evidence and observational studies providing a lower quality of evidence), imprecision of the results over a number of studies, inconsistency of the studies, indirectness, reporting bias, magnitude of the effect, biological plausibility, and strength of association.36-40 • The balance between benefits and harms. Will the intervention do more good than harm? What is the extent of the benefit and of the potential harm? • Feasibility of translating the evidence into the circumstance in which the intervention will be made. Can I apply this in my practice? Is it affordable? • Certainty of the baseline risk. How important is the problem? • Cost. Both monetary costs and expenditures in terms of resources must be considered. The balance between benefit and harm (the trade-off) can be categorized as follows: • Net benefits = the intervention clearly does more good than harm. • Trade-off = there are important trade-offs between the benefits and harms. • Uncertain trade-offs = it is unclear whether the intervention does more good than harm. • No net benefits = the intervention clearly does more harm than benefit. The quality of evidence informing the recommendation can be categorized as follows:

Introduction

VetBooks.ir

• High: We are very confident that the true effect lies close to that of the estimate of the effect. In other words, we can be very confident that both the direction of the effect and its magnitude are known with reasonable certainty and that the magnitude of the effect is clinically relevant. • Moderate: We are moderately confident in the effect estimate. The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different. In other words, the direction of the effect is likely known, although the magnitude might change with further research. The magnitude of the effect is likely clinically significant. • Low: Our confidence in the effect estimate is limited. The true effect could be substantially different from the estimate of the effect. In other words, both the direction of the effect and its magnitude are very likely to change with further research. • Very low: Any estimate of the effect is very uncertain, and the direction and magnitude of the real effect of the intervention are unknown. Finally, all of the considerations just described can be distilled down to the following recommendations being “a judgement that most well-informed people would make”:41 • “Do it”—there is high-quality evidence of net benefits within appropriate resource constraints (costs) for a problem that has significant importance (a judgment that most well-informed people would make). • “Probably do it” = when the strength of evidence is moderate or when the benefit : harm trade-off is unclear or marginal. • “Probably don’t do it” = when the strength of evidence is low or very low, when the benefit : harm trade-off is unclear or marginal, or when the baseline risk is low. • “Don’t do it” = there is high-quality evidence of harm clearly exceeding benefits, the cost is too great compared with benefits, or the baseline risk is very low (i.e., the problem is not important). The GRADE guidelines, although not well established for veterinary medicine, have been used and provide the opportunity to make evidence-based recommendations to practitioners.42

FOOD- AND FIBER-PRODUCING ANIMALS Veterinary practice with food-producing animals provides service primarily to the owners of the meat-, milk-, and fiber-producing animals such as dairy and beef cattle, pigs, sheep, and goats. Veterinarians also provide service to owners of captive ungulates, such as red deer, elk, and bison, which are being raised under farm conditions for the production of meat and byproducts such as hides. Although some commercially processed horsemeat is consumed by humans, the market is small compared with that for beef and pork, and horses are not usually included in discussions about food-producing animal veterinary practice. Poultry, fish, and rabbits are also important sources of human food but are not the subject of this book. For the past several decades, the major activity in food-producinganimal practice, and a major source of income for veterinarians, was the provision of emergency veterinary service to the owners of herds or flocks in which a single animal was affected with one of the common diseases. Occasionally, outbreaks of disease affecting several animals occurred. In addition, routine elective veterinary services, such as castration, vaccination, dehorning, and deworming; the testing for diseases, such as brucellosis and tuberculosis; and the dispensing of veterinary drugs, pharmaceuticals, and biologicals accounted for a significant source of revenue for the veterinarian. Since about the early 1970s, there has been a shift from emphasis and dependence on emergency veterinary medicine and routine procedures to more attention being paid by the veterinarian and the producer to planned animal health and production management using the whole-farm approach. Livestock producers are now much more knowledgeable about animal agriculture and are concerned about the cost-effectiveness and the scientific basis of the recommendations made by veterinarians and agricultural advisors. More and more producers are doing the routine

xv

elective procedures themselves. From firsthand experience and extension courses provided for them, they have also learned how to diagnose and treat many of the common diseases of farm livestock. Many veterinary pharmaceuticals antimicrobials and biologicals can now be purchased by producers from either veterinary or nonveterinary sources.

INDUSTRIALIZED ANIMAL AGRICULTURE The intensification of animal agriculture has created complex animal health and production problems for which there are no simple and reliable therapeutic and preventive procedures, and this has made the task of the veterinarian much more challenging. For example, acute undifferentiated respiratory disease is a common disease of feedlot cattle that is difficult to treat and control effectively because the etiology and epidemiology are complex. Acute diarrhea of calves under 30 days of age may be caused by several different enteropathogens, but a knowledge of the risk factors or epidemiologic determinants, such as colostral immunity and population density, is probably more important for effective clinical management and control of the disease. The rearing of pigs intensively and in complete confinement has exaggerated a number of disease problems, many exacerbated by inadequacies of the environment. Suboptimal reproductive performance resulting from a variety of management and environmental factors is common, and pneumonia in growing and finishing pigs may be almost impossible to eradicate unless the herd is depopulated and repopulated with minimal-disease breeding stock. Infectious diseases such as porcine reproductive and respiratory syndrome are difficult to control. The solutions to these complex problems are not always readily apparent, in part because of insufficient research on etiology and epidemiology and different control strategies in the herds where the problems are occurring. The veterinarian must be knowledgeable and skillful in the principles of epidemiology, applied nutrition, and animal housing; the education and training of animal attendants; and the analysis of production indices, including profit and loss, which includes the use of computers, in addition to being skilled in the traditional veterinary disciplines of medicine, reproduction, pharmacology, and pathology. Thus, the food-producing-animal practitioner must become more skilled in the simultaneous management of animal health and production; the modern livestock producer is cost-conscious, and anything veterinarians do or recommend must be cost-effective.

COMPANION-ANIMAL PRACTICE In contrast, developments in companion-animal medicine (small animals) have followed in the footsteps of human medicine, with an ever-increasing emphasis and reliance on extensive use of clinical pathology for the in-depth evaluation of the hematology, clinical chemistry, enzymology, immune status, and many other body functions of the individual animal. Diagnostic techniques such as ultrasonography, endoscopy, nuclear imaging, and computed tomography are being used both in veterinary teaching hospitals and in referral veterinary practices. These in-depth “diagnostic workups” presumably lead to a greater understanding of the etiology and pathophysiology of disease, with the ultimate aim of a more accurate and early diagnosis that allows much more effective medical and surgical therapy than is economically possible or necessary in food-producing animals. There is not the same emphasis on the efficiency of production, epidemiology, and cost-effectiveness that constantly faces the food-producing-animal practitioner. More and more companion-animal owners, because of the sentimental value of their animals and the growing importance of the human–companion animal bond, are willing to pay for the costs associated with extensive laboratory and sophisticated diagnostic tests and intensive and prolonged veterinary hospital care. Palliative care for dogs and cats affected with diseases that may not be curable over the long term is now a recognized fact in small-animal practice.

xvi

Introduction

VetBooks.ir

EQUINE PRACTICE Equine practice has evolved along similar lines to small-animal practice. Some aspects of it, such as reproduction, intensive clinical care of the newborn foal, and the treatment of medical and surgical diseases of valuable athletic and competitive horses, have advanced a great deal. The great strides that have been made in our understanding of the diagnosis, prognosis, and medical and surgical therapy of colic in the horse are a result of the in-depth diagnostic laboratory work and the medical and surgical expertise that have been used. Our improved understanding of the prognosis of equine colic is in part attributable to prospective studies of the clinical and laboratory findings in horses with colic. However, the large advances in improvement in survival made in the early years of surgical and intensive medical treatment of colic have not continued, and there is an urgent need for appropriately designed prospective clinical trials to determine optimal treatment regimes in these horses. The same is true for intensive treatment of sick foals. In addition to the advanced diagnostic and therapeutic procedures being done on valuable horses at veterinary teaching hospitals, there are now many privately owned equine veterinary centers that provide the same service. Undoubtedly the high financial value of some horses has provided the impetus for the development of these services. Although the increasingly sophisticated diagnostic and therapeutic techniques used in equine practice are readily noted, advances in the understanding of infectious and contagious diseases of horses have also increased markedly. This is particularly true for economically important diseases that have the potential to affect large numbers of horses, consequently causing disruption to important athletic events and the sale and shipment of horses. These diseases are typically the infectious respiratory diseases and those diseases, such as African horse sickness, that are exotic to most of the horse population worldwide. The economic incentive to control these diseases has resulted in considerable increases in knowledge of their etiology (and consequently vaccinology), epidemiology, immunology, diagnosis, and prevention. Few advances have been made in treatment of what are, for the most part, self-limiting diseases with low case-fatality rates.

CONTRASTING OBJECTIVES It is clear that there are major differences between the objectives and principles of companion-animal practice and those of foodproducing-animal practice. In companion-animal practice, the objective is the restoration of the clinically ill animal to a normal state, if possible, or in some cases a less-than-normal state is acceptable provided it is a quality life, using all the readily available diagnostic and therapeutic techniques that can be afforded by the client. In sharp contrast, in food-producing-animal practice, the objective is to improve the efficiency of animal production using the most economical methods of diagnosis, treatment, and control, including the disposal by culling or slaughter of animals that are difficult to treat and are economic losses. This growing dichotomy in the delivery of veterinary services to the food-producing-animal owner and to the companion-animal owner prompted us to present a short introductory commentary on the objectives and principles of food-producing-animal practice.

The Objectives of Food-Producing-Animal Practice EFFICIENCY OF LIVESTOCK PRODUCTION The most important objective in food-producing-animal practice is the continuous improvement of the efficiency of livestock production by the management of animal health. This involves several different but related activities and responsibilities, which include the following:

• Providing the most economical method of diagnosis and treatment of sick and injured animals and returning them to an economically productive status, or to a point where slaughter for salvage is possible, in the shortest possible time. The financially conscious producer wants to know the probability of success following treatment of a disease in an animal and to minimize the costs of prolonged convalescence and repetitive surgery. • Monitoring animal health and production of the herd on a regular basis so that actual performance can be compared with targets and the reasons for the shortfalls in production or increases in the incidence of disease can be identified as soon as possible, so that appropriate and cost-effective action can be taken. The routine monitoring of production records and the regular monitoring of bulk-tank milk somatic cell counts in dairy herds are examples. • Recommending specific disease control and prevention programs, such as herd biosecurity, vaccination of cattle against several important infectious diseases that occur under a variety of conditions, and the strategic use of anthelmintics in cattle and sheep. • Organizing planned herd and flock health programs for the individual farms with the objective of maintaining optimum productivity through animal health management. • Advising on nutrition, breeding, and general management practices. Food-producing-animal practitioners must be interested in these matters when they affect animal health. It is a large part of production-oriented health management, and it is now common for veterinarians to expand their health-oriented animal husbandry advisory service to include an animal-production advisory service. To do so is a matter of individual preference, an option that some veterinarians take up and others do not. Some veterinarians will rely on consultation with agricultural scientists. However, veterinarians still require a working knowledge of the relevant subjects, at least enough to know when to call in the collaborating advisor for advice. Members of both groups should be aware of the extensive list of subjects and speciesoriented textbooks on these subjects, which should be used to support this kind of service.

ANIMAL WELFARE Encouraging livestock producers to maintain standards of animal welfare that comply with the views of the community is emerging as a major responsibility of the veterinarian. The production of foodproducing animals is an animal welfare concern that practitioners face and an area in which they must become proactive.43,44 Increasing public concern for the welfare of animals, including those that produce food and fiber for human consumption, must be addressed using high-quality scientific evidence and a sound understanding of the arguments of individuals and groups opposed to such use of animals.

ZOONOSES AND FOOD SAFETY Promoting management practices that ensure that meat and milk are free of biological and chemical agents capable of causing disease in humans must also become a preoccupation for food-producinganimal veterinarians. This is because the general public is concerned about the safety of the meat and milk products it consumes, and the most effective way to minimize hazards presented by certain infectious agents and chemical residues in meat and milk is to control these agents at their point of entry into the food chain, namely, during the production phase on the farm. Veterinarians will undoubtedly become involved in the surveillance of the use of antimicrobial compounds and other chemicals that are added to feed supplies to promote growth or prevent infections, and they will be expected to minimize the risk

Introduction

VetBooks.ir

of the occurrence of zoonotic disease agents in farm-animal populations.

Principles of Food-Producing Animal Practice REGULAR FARM VISITS A unique feature of a food-producing animal veterinary practice is that most of the service is provided by the veterinarian who makes emergency or planned visits to the farm. In some areas of the world, where veterinarians had to travel long distances to farms, large-animal clinics were established, and producers brought animals that needed veterinary attention to the clinic. For the past 25 years these clinics have provided excellent facilities in which, for example, surgical procedures such as cesarean sections could be done and intensive fluid therapy for dehydrated diarrheic calves could be administered much more effectively and at a higher standard than on the farm. However, much less veterinary service is being provided in these clinics now because of the high operating costs of providing hospital care and the limited economic returns that are possible for the treatment of foodproducing animals, which have a fixed economic value. Producers have also become less enthusiastic about transporting animals to and from a veterinary clinic because of the time and expertise involved, and because of increasing concern about biosecurity and the potential impact of pathogen introduction on the health and productivity of their animals.

CLINICAL EXAMINATION AND DIAGNOSIS The diagnosis, treatment, and control of diseases of food-producing animals are heavily dependent on the results of the clinical examination of animals on the farm and the careful examination of the environment and management techniques. This means that the veterinarian must become highly skilled in obtaining an accurate and useful history on the first visit to an animal or group of animals and in conducting an adequate clinical examination to make the best diagnosis possible, and economically, so that the treatment and control measures can be instituted as soon as possible. On the farm, during the day or in the middle of the night, the veterinarian will not have ready access to a diagnostic laboratory for the rapid determination of a cow’s serum calcium level if milk fever is suspected. The practitioner must become an astute diagnostician and a skillful user of the physical diagnostic skills of visual observation, auscultation, palpation, percussion, succussion, ballottement, and olfactory perception. On the farm, the clinical findings, including the events of the recent disease history of an animal, are often much more powerful, diagnostically, than laboratory data. It therefore becomes increasingly important that the clinical examination should be carefully and thoughtfully carried out so that all clinically significant abnormalities have been detected. An outline of the clinical examinations of an animal and the different methods for making a diagnosis are presented in Chapter 1. Becoming efficient in clinical examination requires the diligent application of a systematic approach to the task and, most importantly, evaluation of the outcome. A most rewarding method of becoming a skillful diagnostician is to retrospectively correlate the clinical findings with the pathology of those cases that die and are submitted for necropsy. The correlation of the clinical findings with the clinical pathology date, if available, is also an excellent method of evaluation but is not routinely available in most private practices. The foodproducing-animal practitioner must also be a competent field pathologist and be able to do a useful necropsy in the field, usually under less-than-desirable conditions, and to make a tentative etiologic diagnosis so that additional cases in the herd can be properly handled or prevented. Doing necropsies on the farm or having them done by a local diagnostic laboratory can be a major activity in a specialty pig or beef feedlot practice, where clinical examination of

xvii

individual animals is done only occasionally, compared with dairy practice.

EXAMINATION OF THE HERD The clinical examination of the herd in which many animals may be affected with one or a number of clinical or subclinical diseases, or in which the owner’s complaint is that performance is suboptimal but the animals appear normal, has become a major and challenging task. This is particularly true in large dairy herds, large pig herds, beef feedlots, lamb feedlots, and sheep flocks where the emphasis is on health management of the herd. Intensified animal agriculture may result in an increased frequency of herd epidemics or outbreaks of diseases such as bovine respiratory disease syndrome, bloat, and acute diarrhea in beef calves and peracute coliform mastitis in dairy cattle. Such well-known diseases are usually recognizable, and a definitive etiologic diagnosis can usually be made, and in some cases the disease can be controlled by vaccination. However, in some cases of herd epidemics of respiratory disease, salmonellosis, or Johne’s disease, for example, the veterinarian may have to make repeated visits to the herd to develop effective treatment and control procedures. The steps involved in the examination of the herd affected by a clinical disease or suboptimal performance are presented in Chapter 1.

COLLECTION AND ANALYSIS OF ANIMAL HEALTH DATA With the shift in emphasis to the problems of the herd, the collection, analysis, and interpretation of animal health and production data will be a major veterinary activity. Livestock producers must keep and use good records if the veterinarian is to make informed decisions about animal health and production. The once tedious and unpopular work of recording and analyzing animal health and production data is now done by the computer. Veterinarians will have to move in the direction of developing a computer-based animal health and production profile of each herd for which they are providing a service. Veterinary colleges will also have to provide leadership and provide undergraduate and graduate student education in the collection, analysis, and interpretation of animal health data. This activity will include methods of informing the producer of the results and the action necessary to correct the herd problem and to improve production.

PUBLIC HEALTH AND FOOD SAFETY Veterinarians have a major responsibility to ensure that the meat and milk produced by the animals under their care are free from pathogens, chemicals, antimicrobials, and other drugs that may be harmful to humans. The prudent use of antimicrobials, including adherence to withdrawal times for meat and milk, are becoming major concerns of the veterinary associations, such as the American Association of Bovine Practitioners, the American Association of Small Ruminant Practitioners, and the American Associate of Swine Practitioners. Traditionally, veterinary public health was not a career option considered by new or recent graduates. However, because of the recent concern about the contamination of meat supplies by pathogens and xenobiotics (any substance foreign to an animal’s biological system), and the potentially serious economic effects of such contamination on the export markets of a country, it is now clear that veterinarians, using a variety of testing techniques, will become increasingly involved in monitoring the use of veterinary drugs so that treated animals are not placed in the food chain until the drugs have been excreted. The same principles apply to the contamination of milk supplies with antimicrobials, prevention of which is a major responsibility of the veterinarian.

ECONOMICS OF VETERINARY PRACTICE The successful delivery of food-producing-animal practice will depend on the ability of the veterinarian to provide those services that

xviii

Introduction

VetBooks.ir

the producer needs and wants at a price that is profitable to both the producer and veterinarian. Several constraints interfere with this successful delivery. Maximizing net profit is not a high priority for many farmers. Being independent and making a living on the farm are commonly ranked higher. Consequently, when veterinarians make recommendations to control a disease, their subsequent enthusiasm for giving advice may be dampened if farmers do not adopt the control procedures even though the advice is based on good information about expected economic returns. The frustrations that many veterinarians experience in attempting to get dairy producers to adopt the principles of an effective and economical mastitis control program are well known. In some cases, producers do not use modern methods of production and disease control because they are unaware of their importance. The variable financial returns that farmers receive for their commodities, particularly the low prices received during times of oversupply of meat and milk, may also influence whether they purchase professional veterinary service or attempt to do the work themselves.

VETERINARY EDUCATION We have described our views on the state of food-producing-animal medicine and what it requires of veterinarians who practice it. Traditionally, veterinary colleges have provided undergraduate students with the knowledge and clinical skills necessary to enter veterinary practice and begin to engage in food-producing-animal practice. Fieldservice units and large-animal in-clinics devoted to clinical teaching have been an integral part of most veterinary colleges. The clinical caseload is for the students, clinicians, and those in the paraclinical sciences such as microbiology, toxicology, clinical pathology, and pathology. However, recently, it seems that veterinary colleges have not maintained their farm-animal teaching clinics, and in fact, some of these teaching clinics have ceased to exist. The demise of in-house food-producing-animal practice in veterinary teaching hospitals, as opposed to the care of agricultural animals from hobby farms, is contributed to by the increasing use of stringent biosecurity measures on medium- and large-scale operations. Animals brought to veterinary teaching hospitals for diagnosis and possible treatment cannot be returned to the farm because of the fear of introducing infectious disease. Regardless, the demise of in-house food-producing-animal practice in some universities should be of major concern to the veterinary profession because universities have an obligation to serve the veterinary needs of animal agriculture. Some veterinary colleges have developed extensive programs in which undergraduate students spend time in private veterinary practice to gain clinical experience. However, the failure to maintain and support viable farm-animal teaching clinics will diminish the clinical experience of clinicians and those in the paraclinical sciences who have a primary responsibility for teaching. In addition, the lack of clinical cases will adversely affect the clinical research activities of clinicians. Clinicians must experience a critical number of clinical cases to maintain credibility as a veterinary scholar. To study the phenomena of disease without books is to sail an unchartered sea, while to study books without patients is not to go to sea at all. Sir William Osler (Books and Men, Boston Surgical Journal, 1901) The practicing veterinarian must become knowledgeable about various aspects of farm animal management, especially those that cause or contribute to clinical or subclinical disease and impaired animal production. Such veterinarians will become species-industry specialists who can provide totally integrated animal health and production management advice to those managing a dairy herd, a beef cow-calf herd, a beef feedlot, a pig herd, or a sheep flock. To be able to do this, veterinarians will need to undertake a postgraduate clinical residency program or develop the expertise on their own by diligent self-education in a veterinary practice that is committed to the concept of a total animal health management and allows the veterinarian the time and the resources to develop the specialty.

OPTIMAL UTILIZATION OF THE FOODPRODUCING-ANIMAL PRACTITIONER All that we have said in this Introduction is related to enhancing and improving the performance of the professional food-producinganimal veterinarian. In developed countries, this could mean greater utilization of each veterinarian by farmers and improved financial viability of their farming enterprises. In developing countries, it could mean a greater volume of production at a time when malnutrition appears to be the fate of so many groups of the world community. These could be the outcomes if the world’s agricultural situation was a stable one. As it is, there is currently a great upheaval in agriculture; developed countries are heavily overproduced, and there is a sharp decline in farming as an industry and way of life. In developing countries, the decisions governing the health and welfare of animals and the people that depend on them often seem to depend more on political expediency than on the basic needs of humans and their animals. In these circumstances we do not feel sufficiently courageous and farsighted to predict our individual futures, but with the hindsight of how far the human population and the attendant agricultural and veterinary professions have come in the past 56 years, we are confident that you will have an opportunity to properly pursue the objectives and principles that we have described. FURTHER READING

Animal agriculture in a changing climate. Cornell University. . Centre for Evidence-Based Veterinary Medicine. University of Nottingham. . Quammen D. Spillover: Animal Infections and the Next Human Epidemic. London: Vintage Books; 2013. Thornton PK, van de Steeg J, Notenbaert A, et al. The impacts of climate change on livestock and livestock systems in developing countries: a review of what we know and what we need to know. Ag Syst. 2009;101:113-127.

REFERENCES

1. Crutzen PJ. Nature. 2002;415:23. 2. Whitmee S, et al. Lancet. 2015;386:1973. 3. Gauly M, et al. Animal. 2013;7:843. 4. Thornton PK, et al. Ag Syst. 2009;101:113. 5. Keesing F, et al. Nature. 2010;468:647. 6. Plowright RK, et al. Proc Royal Soc B. 2015;282. 7. Plowright RK, et al. Proc Royal Soc B. 2011;278:3703. 8. Pulliam JRC, et al. J R Soc Interface. 2012;9:89. 9. Gould EA, et al. Trans R Soc Trop Med Hyg. 2009;103:109. 10. MacLachlan NJ, et al. Vet Res. 2010;41. 11. Maclachlan NJ, et al. Rev Sci Tech. 2015;34:329. 12. Wilson A, et al. Parasitol Res. 2008;103:S69. 13. Jacquet S, et al. Mol Ecol. 2015;24:5707. 14. Paweska JT. Rev Sci Tech. 2015;34:375. 15. Doceul V, et al. Vet Res. 2013;44. 16. Gale P, et al. J Appl Microbiol. 2009;106:1409. 17. Thompson GM, et al. Ir Vet J. 2012;65:(3 May 2012). 18. Faverjon C, et al. BMC Vet Res. 2015;11. 19. Webster WR. Aust Vet J. 2011;89:3. 20. Smyth GB, et al. Aust Vet J. 2011;89:151. 21. Roeder P, et al. Philos Trans R Soc Lond B Biol Sci. 2013;368. 22. Cool cows: dealing with heat stress in Australian dairy herds. Dairy Australia, 2016. Accessed May 1, 2016, at . 23. Animal agriculture in a changing climate. Cornell University, 2016. Accessed May 1, 2016, at . 24. Holmes M, et al. In Pract. 2004;26:28. 25. Cockcroft P, et al. In Pract. 2004;26:96. 26. Holmes M, et al. In Pract. 2004;26:154. 27. Evidence-based veterinary medicine. University of Nottingham. Accessed April 2, 2016, at . 28. Sargeant JM, et al. Zoonoses Pub Health. 2014;61:10. 29. O’Connor A, et al. Vet J. 2015;206:261. 30. O’Connor AM, et al. Zoonoses Pub Health. 2014;61:28. 31. O’Connor AM, et al. Zoonoses Pub Health. 2014;61:52. 32. Sargeant JM, et al. Zoonoses Pub Health. 2014;61:39. 33. Sargeant JM, et al. J Vet Intern Med. 2010;24:44.

Introduction

VetBooks.ir

34. O’Connor AM, et al. J Vet Intern Med. 2010;24:57. 35. Guyatt G, et al. J Clin Epidemiol. 2011;64:383. 36. Guyatt GH, et al. J Clin Epidemiol. 2011;64:1283. 37. Guyatt GH, et al. J Clin Epidemiol. 2011;64:1303. 38. Guyatt GH, et al. J Clin Epidemiol. 2011;64:1294. 39. Guyatt GH, et al. J Clin Epidemiol. 2011;64:1311.

40. Guyatt GH, et al. J Clin Epidemiol. 2011;64:407. 41. Guyatt GH, et al. Br Med J. 2008;336:1049. 42. Hinchcliff KW, et al. J Vet Intern Med. 2015;29:743. 43. Coetzee JF. Appl Anim Behav Sci. 2011;135:192. 44. Marley CL, et al. Animal. 2010;4:259.

xix

This page intentionally left blank

VetBooks.ir

VetBooks.ir

Contents Contributors,  ix Preface to the Eleventh Edition,  x Introduction,  xii List of Tables,  xxiv List of Illustrations,  xxvii

1 Clinical Examination and Making a Diagnosis,  1 Introduction,  1 Making a Diagnosis,  2 Clinical Examination of the Individual Animal,  5 Prognosis and Therapeutic Decision Making,  26

2 Examination of the Population,  29 Approach to Examining the Population,  29 Examination Steps,  30 Techniques in Examination of the Herd or Flock,  32 Role of the Integrated Animal Health and Production Management Program,  34

3 Biosecurity and Infection Control,  36 Definitions and Concepts,  36 Development of a Biosecurity   Plan,  37 Practices to Aid in Maintaining Biosecurity,  38

4 General Systemic States,  43 Hypothermia, Hyperthermia, and Fever,  43 Acute Phase Response,  56 Sepsis, Septicemia, and Viremia,  57 Toxemia, Endotoxemia, and Septic Shock,  59 Toxemia in the Recently Calved Cow,  67 Hypovolemic, Hemorrhagic, Maldistributive, and Obstructive Shock,  71 Localized Infections,  76 Pain,  78 Stress,  84 Disturbances of Appetite, Food Intake, and Nutritional Status,  87 Weight Loss or Failure to Gain Weight (Ill-Thrift),  90 Physical Exercise and Associated Disorders,  96 Sudden or Unexpected Death,  99 Diseases Associated With Physical Agents,  103 Diagnosis of Inherited Disease,  111

5 Disturbances of Free Water, Electrolytes, Acid-Base Balance, and Oncotic Pressure,  113 Dehydration,  113 Water Intoxication,  115 Electrolyte Imbalances,  116 Acid-Base Imbalance,  123 Oncotic Pressure and Edema,  128 Naturally Occurring Combined Abnormalities of Free Water, Electrolyte, Acid-Base Balance, and Oncotic Pressure,  130 Principles of Fluid and Electrolyte Therapy,  137

6 Practical Antimicrobial Therapeutics,  153 Principles of Antimicrobial   Therapy,  153 Antibiotic Resistance,  156 Antibiotic Metaphylaxis to Control Respiratory Disease,  158 Practical Usage of Antimicrobial Drugs,  158 Classification of Antimicrobial Agents: Mechanisms of Action and Major Side Effects,  169 β-Lactam Antibiotics: Penicillins, Cephalosporins, and β-Lactamase Inhibitors,  170

7 Diseases of the Alimentary Tract: Nonruminant,  175 Principles of Alimentary Tract Dysfunction,  176 Manifestations of Alimentary Tract Dysfunction,  178 Special Examination,  183 Principles of Treatment in Alimentary Tract Disease,  190 Diseases of the Buccal Cavity and Associated Organs,  192 Diseases of the Pharynx and Esophagus,  196 Diseases of the Nonruminant Stomach and Intestines,  203 Diseases of the Peritoneum,  215 Abdominal Diseases of the Horse Including Colic and Diarrhea,  220 Abdominal Diseases of the Pig Including Diarrhea,  287 Noninfectious Intestinal Disease of Swine,  290 Bacterial and Viral Diseases of the Alimentary Tract,  292 Parasitic Diseases of the Alimentary Tract,  397 Control,  421 Toxins Affecting the Alimentary Tract,  421 Neoplasms of the Alimentary Tract,  431

Congenital Defects of the Alimentary Tract,  432 Inherited Defects of the Alimentary Tract,  434

8 Diseases of the Alimentary Tract–Ruminant,  436 Diseases of the Forestomach of Ruminants,  436 Special Examination of the Alimentary Tract and Abdomen of Cattle,  445 Diseases of the Rumen, Reticulum and Omasum,  457 Diseases of the Abomasum,  500 Diseases of the Intestines of Ruminants,  523 Bacterial Diseases of the Ruminant Alimentary Tract,  531 Viral Diseases of the Ruminant Alimentary Tract,  572 Parasitic Diseases of the Ruminant Alimentary Tract,  603 Toxic Diseases of the Ruminant Alimentary Tract,  618 Diseases of the Ruminant Alimentary Tract of Unknown Cause,  621

9 Diseases of the Liver,  622 Diseases of the Liver: Introduction,  622 Principles of Hepatic Dysfunction,  622 Manifestations of Liver and Biliary Disease,  623 Special Examination of the Liver,  625 Principles of Treatment in Diseases of the Liver,  629 Diffuse Diseases of the Liver,  629 Diseases Characterized by Systemic Involvement,  639 Hepatic Diseases Associated With Trematodes,  641 Diseases Associated With Major Phytotoxins,  645 Poisoning by Mycotoxins,  649 Focal Diseases of the Liver,  655 Diseases of the Pancreas,  656

10 Diseases of the Cardiovascular System,  657 Principles of Circulatory Failure,  657 Manifestations of Circulatory Failure,  659 Special Examination of the Cardiovascular System,  663 Arrhythmias (Dysrhythmias),  675 Diseases of the Heart,  685 Cardiac Toxicities,  697 Cardiac Neoplasia,  703 Congenital Cardiovascular Defects,  703 Inherited Defects of the Circulatory System,  706 Diseases of the Pericardium,  707 Diseases of the Blood Vessels,  709 Vascular Neoplasia,  715 xxi

xxii

Contents

VetBooks.ir

11 Diseases of the Hemolymphatic and Immune Systems,  716 Abnormalities of Plasma Protein Concentration,  716 Hemorrhagic Disease,  718 Lymphadenopathy (Lymphadenitis),  751 Diseases of the Spleen and Thymus,  752 Immune-Deficiency Disorders (Lowered Resistance to Infection),  753 Amyloidoses,  755 Enzootic Bovine Leukosis (Bovine Lymphosarcoma),  785 Nutritional Deficiencies,  814 Toxins Affecting the Hemolymphatic System,  823 Neoplasia,  834 Congenital Inherited Diseases,  837 Inherited Immunodeficiency,  839 Diseases of Unknown Etiology,  842

12 Diseases of the Respiratory System,  845 Principles of Respiratory Insufficiency,  846 Principal Manifestations of Respiratory Insufficiency,  848 Special Examination of the Respiratory System,  855 Principles of Treatment and Control of Respiratory Tract Disease,  868 Diseases of the Upper Respiratory Tract,  874 Diseases of the Lung   Parenchyma,  880 Diseases of the Pleural Cavity and Diaphragm,  895 Diseases of the Bovine Respiratory Tract,  901 Diseases of the Ovine and Caprine Respiratory Tract,  969 Diseases of the Equine Respiratory Tract,  981 Diseases of the Swine Respiratory Tract,  1047 Respiratory System Toxicoses,  1087 Neoplastic Diseases of the Respiratory Tract,  1088 Congenital and Inherited Diseases of the Respiratory Tract,  1090

13 Diseases of the Urinary System,  1095 Introduction,  1095 Clinical Features of Urinary Tract Disease,  1097 Special Examination of the Urinary System,  1099 Principles of Treatment of Urinary Tract Disease,  1108 Diseases of the Kidney,  1110 Infectious Diseases of   the Kidney,  1115 Toxic Agents Affecting   the Kidney,  1135 Renal Neoplasia,  1137 Congenital and Inherited Renal Diseases,  1137

Diseases of the Ureters, Bladder, and Urethra,  1139 Diseases of the Prepuce and Vulvovaginal Area,  1152

14 Diseases of the Nervous System,  1155 Introduction,  1156 Principles of Nervous   Dysfunction,  1157 Clinical Manifestations of Diseases of the Nervous System,  1158 Special Examination of the Nervous System,  1164 Diffuse or Multifocal Diseases of the Brain and Spinal Cord,  1178 Focal Diseases of the Brain and Spinal Cord,  1189 Plant Toxins Affecting the Nervous System,  1194 Fungal Toxins Affecting the Nervous System,  1201 Other Toxins Affecting the Nervous System,  1202 Diseases of the Cerebrum,  1219 Bacterial Diseases Primarily Affecting the Cerebrum,  1224 Viral Diseases Primarily Affecting the Cerebrum,  1227 Prion Diseases Primarily Affecting the Cerebrum,  1286 Parasitic Disease Primarily Affecting the Cerebrum,  1301 Metabolic Diseases Primarily Affecting the Cerebrum,  1302 Metabolic and Toxic Encephalomyelopathies,  1321 Inherited Diseases Primarily Affecting the Cerebrum,  1322 Congenital and Inherited Encephalomyelopathies,  1324 Diseases Primarily Affecting the Cerebellum,  1328 Diseases Primarily Affecting the Brainstem and Vestibular System,  1329 Diseases Primarily Affecting the Spinal Cord,  1337 Parasitic Diseases Primarily Affecting the Spinal Cord,  1341 Toxic Diseases Primarily Affecting the Spinal Cord,  1346 Inherited Diseases Primarily Affecting the Spinal Cord,  1346 Diseases Primarily Affecting the Peripheral Nervous System,  1358

15 Diseases of the Musculoskeletal System,  1371 Principal Manifestations of Musculoskeletal Disease,  1372 Diseases of Muscles,  1377 Diseases of Bones,  1388 Diseases of Joints,  1406 Infectious Diseases of the Musculoskeletal System,  1425 Nutritional Diseases Affecting the Musculoskeletal System,  1458

Toxic Agents Affecting the Musculoskeletal System,  1503 Congenital Defects of Muscles, Bones, and Joints,  1510 Inherited Diseases of Muscles,  1514 Inherited Diseases of Bones,  1530 Inherited Diseases of Joints,  1538

16 Diseases of the Skin, Eye, Conjunctiva, and External Ear,  1540 Introduction,  1541 Principles of Treatment of Diseases of the Skin,  1543 Diseases of the Epidermis and Dermis,  1543 Diseases of the Hair, Wool, Follicles, and Skin Glands,  1552 Diseases of the Subcutis,  1555 Non-Infectious Diseases of the Skin,  1559 Bacterial Diseases of the Skin,  1564 Viral Diseases of the Skin,  1580 Dermatomycoses,  1600 Protozoal Diseases of the Skin,  1607 Nematode Infections of   the Skin,  1608 Cutaneous Myiasis,  1611 Mite Infestations,  1618 Ked and Louse Infestations,  1623 Miscellaneous Skin Diseases Caused   by Flies, Midges, and Mosquitoes,  1625 Tick Infestations,  1631 Deficiencies and Toxicities Affecting the Skin,  1634 Cutaneous Neoplasms,  1640 Congenital and Inherited Defects of the Skin,  1643 Eye and Conjunctival Diseases,  1648 External Ear Diseases,  1660

17 Metabolic and Endocrine Diseases,  1662 Introduction,  1662 Metabolic Diseases of   Ruminants,  1662 Inherited Metabolic Diseases of Ruminants,  1727 Metabolic Diseases of Horses,  1727 Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma),  1739 Diseases Caused by Nutritional Deficiencies,  1747 Deficiencies of Energy and Protein,  1753 Diseases Associated with Deficiencies of Mineral Nutrients,  1754

18 Diseases Primarily Affecting the Reproductive System,  1758 Infectious Diseases Primarily   Affecting the Reproductive System,  1758 Infectious Diseases Primarily Affecting the Reproductive System,  1761

Contents

VetBooks.ir

Toxic Agents Primarily Affecting the Reproductive System,  1821 Congenital and Inherited Diseases Primarily Affecting the Reproductive System,  1828

19 Perinatal Diseases,  1830 Introduction,  1830 Perinatal and Postnatal Diseases,  1830 Perinatal Disease—Congenital Defects,  1835 Physical and Environmental Causes of Perinatal Disease,  1840 Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin),  1848 Clinical Assessment and Care of Critically Ill Newborns,  1856 Neonatal Infectious Diseases,  1874 Neonatal Neoplasia,  1903

20 Diseases of the Mammary Gland,  1904 Introduction,  1904 Bovine Mastitis,  1904 Diagnosis of Bovine Mastitis,  1914 Mastitis Pathogens of Cattle,  1930

Mastitis of Cattle Associated With Common Contagious Pathogens,  1930 Mastitis of Cattle Associated With Teat Skin Opportunistic Pathogens,  1942 Mastitis of Cattle Associated With Common Environmental Pathogens,  1943 Mastitis of Cattle Associated With Less Common Pathogens,  1960 Control of Bovine Mastitis,  1964 Miscellaneous Abnormalities of the Teats and Udder,  1985 Mastitis of Sheep,  1991 Mastitis of Goats,  1993 Contagious Agalactia in Goats and Sheep,  1994 Mastitis of Mares,  1996 Postpartum Dysgalactia Syndrome of Sows,  1996

21 Systemic and Multi-Organ Diseases,  2002 Diseases of Complex or Undetermined Etiology,  2003 Multi-Organ Diseases Due to Bacterial Infection,  2011

xxiii

Multi-Organ Diseases Due to Viral Infection,  2058 Multi-Organ Diseases Due to Protozoal Infection,  2137 Multi-Organ Diseases Due to Trypanosome Infection,  2150 Multi-Organ Diseases Due to Fungal Infection,  2158 Multi-Organ Diseases Due to Metabolic Deficiency,  2161 Multi-Organ Diseases Due to Toxicity,  2176

APPENDICES,  2215 Appendix 1  Conversion Tables,  2215 Appendix 2   Reference Laboratory Values,  2217 Appendix 3  Drug doses and intervals for horses and ruminants,  2220 Appendix 4  Drug doses and intervals for pigs,  2232

Index,  2235

VetBooks.ir

List of Tables 1-1 1-2 1-3

1-4

3-1

3-2

3-3

4-1

5-1

5-2

5-3

5-4 5-5

5-6

6-1

7-1

7-2

7-3

xxiv

Normal average temperatures with critical points,  15 Resting pulse rates,  15 Method for determining sensitivity, specificity, likelihood ratio for positive and negative tests, positive predictive value, and negative predictive value of a test,  24 Effect of changes in prevalence (pretest probability of disease) on the positive predictive value and negative predictive value of tests with 95% sensitivity and specificity (Test A) and 60% sensitivity and specificity (Test B),  25 Examples of online risk calculators that can help veterinarians and producers estimate the risk of introducing diseases onto facilities in which animals are maintained,  36 Examples of online sources of information regarding biosecurity practices appropriate for agricultural animal species,  36 Examples of some classes of disinfectants with some of their benefits and drawbacks,  40 A simple method of categorizing livestock affected by burns and appropriate actions for each category,  111 Representative laboratory values (mean ± sd) in body water and electrolyte disturbances,  133 Summary of disturbances of body water, electrolytes, and acid-base balance in some common diseases of cattle and horses, and suggested fluid therapy,  137 Summary of effective strong ion difference and osmolarity of parenterally administered crystalloid solutions,  138 Estimated daily energy requirements of fasting cattle,  140 Composition (mmol/L) and indications for use of electrolyte solutions used in fluid therapy,  144 Examples of approximate amounts of fluid required for rehydration and maintenance therapy,  145 The four major categories of penicillin: Narrow spectrum, broad spectrum, penicillinase resistant, and extended spectrum,  170 Medications with potential prokinetic actions in horses (see text for references),  181 Guidelines for the classification and interpretation of bovine peritoneal fluid,  187 Characteristics of equine peritoneal fluid in selected diseases of horses,  187

7-4

7-5 7-6

7-7

7-8 7-9 7-10 7-11

7-12 7-13 7-14 7-15 7-16

7-17 7-18 7-19 7-20

7-21

7-22 7-23 7-24

7-25

7-26 7-27

7-28

Epidemiological and clinical features of diseases of cattle in which diarrhea is a significant clinical finding,  205 Epidemiological and clinical features of horses with diarrhea,  206 Epidemiological and clinical features of diseases of the pig in which diarrhea is a significant clinical finding,  206 Epidemiological and clinical features of the diseases of sheep and goats in which diarrhea is a significant clinical finding,  207 Fluid deficits (L) in horses, foals and calves with 10% dehydration,  213 Origin and examples of visceral pain in the horse,  221 Etiological classification of equine colic,  222 Disorders of the equine gastrointestinal tract causing colic, by anatomical site,  222 Criteria for evaluation of pain in horses,  226 Rectal findings and associated causes of equine colic,  228 Method for brief examination of the abdomen of a horse with colic,  230 Analgesics and spasmolytics for use in equine colic,  233 Promotility agents, lubricants, and fecal softeners for use in horses with colic,  235 Diseases causing colic in foals,  238 Differential diagnosis of common foal colics,  240 Differential diagnosis of common equine colics,  242 Drugs used in the treatment of clinically important gastroduodenal ulcer disease of foals and adult horses and recommendations for treatment (not prophylaxis),  246 Agents used to treat ileus in horses with duodenitis and proximal jejunitis,  257 Epidemiological and clinical features of suckling foals with diarrhea,  274 Proportion of samples testing positive for one of the listed organisms,  294 Biochemical characteristics of species of Brachyspira isolated from pigs,  327 Importance of fomites and animals in spread of Brachyspira sp. on one farm,  328 Antibiotic therapy in use for swine dysentery,  332 Differential diarrhea: Most likely causes of acute neonatal diarrhea in farm animals,  373 Calf diarrhea risk factors: Risk factors and their role in acute undifferentiated diarrhea of newborn calves,  374

7-29 Calf enteropathogens: Age occurrence of the common enteropathogens in calves,  376 7-30 Chemotherapeutics recommended for treatment and control of coccidiosis in calves and lambs,  406 8-1 Effects of some common clinical excitatory and inhibitory influences on primary cycle movements of the reticulorumen,  438 8-2 Differential diagnosis of causes of gastrointestinal dysfunction of cattle,  442 8-3 Differential diagnosis of abdominal distension in cattle,  447 8-4 Pathogenesis and interpretation of clinical findings associated with diseases of the digestive tract and abdomen of cattle,  454 8-5 Diseases of the digestive tract and abdomen of cattle in which a laparotomy is indicated if the diagnosis can be made,  455 8-6 Clinical and laboratory indications for an exploratory laparotomy in cattle when the diagnosis is not obvious,  455 8-7 Differential diagnosis of diseases of the digestive tract and abdomen of young calves presented with distension of the abdomen,  456 8-8 Guidelines for the use of clinical findings in assessing the severity of grain overload in cattle for the selection of the treatment of choice,  466 8-9 The relationship between the stages in the pathogenesis of Johne’s disease, the presence of clinical disease and the results of diagnostic test,  557 8-10 Differential diagnosis of diseases of cattle in which there are either oral lesions or diarrhea alone or together in the same animal,  590 8-11 Anatomical distribution of trichostrongylid worms in ruminants,  604 10-1 Base–apex electrocardiographic parameters in cattle and horses (mean ± standard deviation),  668 10-2 Mean (± standard deviation) cardiopulmonary values for adult horses, cattle and calves, and pigs,  674 10-3 Results of sequential blood gas analysis from a 2-year-old Holstein-Friesian heifer with a large ventricular septal defect,  674 10-4 Common arrhythmias and conduction disturbances in the horse and cow,  675

List of Tables

VetBooks.ir

10-5 Evaluation of pulmonary arterial pressure scores in cattle examined at high altitude,  695 11-1 Differential diagnosis of anemia, with or without edema, in horses,  729 11-2 Differential diagnosis of diseases of cattle characterized by acute hemolytic anemia with or without hemoglobinuria,  730 11-3 Characteristic or expected changes in hematological and serum biochemical variables in anemic animals,  733 11-4 Acute hemorrhage: estimated blood loss,  735 11-5 Method for performing the jaundiced foal agglutination test,  744 11-6 Protocols for treatment of lymphoma and lymphosarcoma in adult equids,  749 11-7 Defects of acquired immunity causing disease in foals and horses,  754 11-8 Algorithm for testing horses for infection by equine infectious anemia virus,  798 11-9 Ticks reported to transmit protozoan disease,  800 11-10 Major Babesia species infective to domestic animals, their tick vectors, and their geographic distribution,  801 11-11 Differential diagnosis of diseases of cattle in which red urine is a principal manifestation,  807 12-1 Identification and clinical significance of breath sounds,  851 12-2 Guidelines for radiographic pulmonary pattern recognition in foals,  858 12-3 Representative results of cytology of bronchoalveolar lavage fluid of cattle, sheep, pigs, and horses,  863 12-4 Changes in blood gas tensions in various disease states compared with values in normal animals breathing air at sea level,  866 12-5 Effects of unilateral or bilateral nasal insufflation of oxygen at flow rates of 50 mL O2 per kg bodyweight per minute to 5- to 7-day-old healthy foals on inspired oxygen tension (measured in the thoracic trachea), arterial oxygen tension and measures of acid : base balance. (Reproduced from Wong et al.2010.),  870 12-6 Major pathogenic Mycoplasma spp. of ruminants, swine, and horses,  885 12-7 Differential diagnosis of bovine respiratory disease,  904 12-8 Contagious pleuropneumonia-1: Members of the Mycoplasma mycoides cluster,  926 12-9 Summary of systemic mycoplasmoses of sheep and goats,  971 12-10 Preferred terms for describing findings on endoscopic examination of the upper airway of horses (modified from),  982 12-11 Antimicrobial agents and recommended doses for treatment of pleuropneumonia in horses,  994

12-12 Causes of epistaxis in horses,  1001 12-13 Drugs used in the treatment of heaves in horses,  1010 12-14 Frequency and prevalence of extrapulmonary manifestations of infection by R. equi in 150 foals. (Reproduced with permission.),  1015 12-15 Differential diagnosis of respiratory diseases of older (not newborn) foals,  1017 12-16 Differential diagnosis of diseases of the upper respiratory tract of horses,  1023 12-17 Aims and associated measures used to control transmission of Streptococcus equi in affected premises and herds,  1025 13-1 Indices of renal function in healthy adult horses and foals less than 30 days of age,  1106 13-2 Forms of leptospirosis in the animal species,  1120 13-3 Differential diagnosis of diseases of cattle characterized by acute hemolytic anemia with or without hemoglobinuria,  1126 14-1 Correlation between clinical findings and location of lesions in the nervous system of farm animals: abnormalities of mental state (behavior),  1165 14-2 Correlation between clinical findings and location of lesion in the nervous system of farm animals: involuntary movements,  1166 14-3 Correlation between clinical findings and location of lesion in the nervous system of farm animals: abnormalities of posture,  1166 14-4 Correlation between clinical findings and location of lesion in the nervous system of farm animals: abnormalities of gait,  1167 14-5 Correlation between clinical findings and location of lesion in the nervous system of farm animals: abnormalities of the visual system,  1168 14-6 Correlation between clinical findings and location of lesion in the nervous system of farm animals: disturbances of prehension, chewing, or swallowing,  1168 14-7 Needle length gauge for lumbosacral cerebrospinal fluid collection,  1173 14-8 Relative diffusion of gram-negative antimicrobials,  1183 14-9 Diseases of horses characterized by signs of intracranial or disseminated lesions of the central nervous system,  1233 14-10 Differential diagnosis of diseases of cattle with clinical findings referable to brain dysfunction,  1235 14-11 Viruses causing encephalomyelitis in horses. reproduced with permission.,  1259 14-12 Commercial vaccines against alphaviral equine encephalomyelitis available for equines,  1272

xxv

14-13 Three-tiered approach to managing an outbreak of equine herpesvirus myeloencephalopathy.,  1281 14-14 Transmissible spongiform encephalopathies in animals and humans,  1286 14-15 PrP genotype and susceptibility to scrapie in national scrapie program in Great Britain,  1297 14-16 Scrapie susceptibility and genotype as defined by the U.S. Scrapie Eradication Plan,  1297 14-17 Daily dietary allowances of vitamin A,  1320 14-18 Diagnostic taxonomy of congenital tremor in pigs,  1327 14-19 Key features of the six types of congenital tremor described in pigs,  1327 14-20 Association of horse factors associated with a diagnosis of cervical stenotic myelopathy in 811 horses with cervical stenotic myelopathy and 805 control horses,  1352 14-21 Differential diagnosis of disease causing spinal ataxia in adult horses,  1356 14-22 Ticks reported to cause paralysis in livestock,  1367 15-1 Differential diagnosis of diseases of the musculoskeletal system,  1373 15-2 Common or well-characterized myopathies of equids,  1383 15-3 Laboratory evaluation of synovial fluid in diseases of the joints,  1409 15-4 Differential diagnosis of lameness accompanied by foot lesions in sheep,  1445 15-5 Virulence of strains,  1453 15-6 Antimicrobial resistance in Spain and the UK.,  1454 15-7 Glutathione peroxidase (GSH-PX) activity and selenium concentration in blood and body tissues of animals deficient in selenium,  1472 15-8 Selenium reference range to determine selenium status of sheep and cattle in New Zealand,  1472 15-9 Examples of estimated daily requirements of calcium, phosphorus, and vitamin D,  1486 15-10 Inherited skeletal diseases of livestock with known mutations and proposed mechanisms.,  1531 15-11 Differential diagnoses for inherited skeletal diseases of animals,  1533 16-1 Terms used to identify skin lesions,  1542 16-2 Differential diagnosis of diseases of swine with skin lesions,  1578 16-3 Differential diagnosis of diseases of horses characterized by discrete lesions of the skin only,  1598 16-4 Differential diagnosis of diseases of horses characterized by lesions of the skin of the lower limbs only,  1599 16-5 Single- and multiple-host ticks,  1631

xxvi

List of Tables

VetBooks.ir

16-6 Diseases associated with bacteria, viruses, and rickettsia and reported to be transmitted by ticks,  1632 17-1 Salient features of metabolic diseases of farm animals,  1663 17-2 Metabolic profile parameters in cattle—optimum values,  1672 17-3 Annual (April–March) percentages outside optimum ranges of metabolite results in blood plasma in adult dairy cows,  1673 17-4 Relationship between the 10-point BCS scale used in New Zealand and the 5-point BCS scale used in Ireland and the United States, and the 8-point scale used in Australia,  1675 17-5 Parturient paresis: Differential diagnosis of common causes of recumbency in parturient adult cattle,  1682 17-6 Parturient paresis: Molecular weights, equivalent weights, and conversions from percent to milliequivalents (%–mEq) of anions and cations used in calculating dietary cation–anion difference,  1687 17-7 To convert from the SI unit to the conventional unit, divide by the conversion factor; to convert from the conventional unit to the SI unit, multiply by the conversion factor,  1712 17-8 Diagnostic Testing Methods for Equine pituitary pars intermedia dysfunction (PPID),  1729 17-9 Criteria for estimating body condition in light-breed horses,  1733 17-10 Grading system for assessing regional adiposity in the neck of ponies and horses,  1733 17-11 Serum or plasma concentrations of thyroid hormones and thyroidstimulating hormone in foals, horses, donkeys, and cattle,  1741 17-12 Principal pathologic and metabolic defects in essential trace-element deficiencies,  1755 18-1 Diagnostic summary of infectious abortion in ewes,  1777 18-2 Diagnostic summary of causes of abortion in cattle,  1788 18-3 Cytokines and porcine reproductive and respiratory syndrome,  1802

18-4 Nursey depopulation and cleanup protocol for elimination of PRRS,  1811 19-1 Scoring system for assessing vigorousness of newborn lambs,  1848 19-2 Definitions for lamb behaviors,  1848 19-3 Failure of transfer of passive immunity; concentrations and relative percentage of immunoglobulins in serum and mammary secretions of cattle and pigs,  1849 19-4 Criteria to assess stage of maturity of the newborn foal,  1859 19-5 Hematologic values of normal foals and calves,  1861 19-6 Serum biochemical values of normal foals and calves,  1862 19-7 Antimicrobials used in neonatal foals,  1866 19-8 Method for calculating survival score in hospitalized neonatal foals,  1867 19-9 Probability of survival for hospitalized neonatal foals with survival scores calculated according to table 19-8,  1867 19-10 Differential diagnosis of comatose (“sleeper”) neonatal foals,  1873 19-11 Worksheet for calculating a sepsis score for foals less than 12 days of age,  1877 19-12 Degree of dehydration in calves with experimentally induced diarrhea,  1886 19-13 Possible causes of bacteremia/ septicemia and acute neonatal diarrhea in farm animals,  1888 20-1 Estimated prevalence of infection and losses in milk production associated with bulk tank milk somatic cell count,  1915 20-2 Sensitivities and specificities for diagnosing an intramammary infection based on the culture of a single milk sample from a quarter using a 10-µL volume,  1916 20-3 Calculating somatic cell score (previously called linear score) from the somatic cell count,  1918 20-4 Conversion of somatic cell scores (previously called linear scores) to somatic cells counts (cells/mL) and predicted loss of milk,  1918

20-5 California mastitis test reactions and equivalent somatic cell counts and somatic cell scores for bovine milk and somatic cell counts for bovine colostrum,  1918 20-6 Summary of three-compartment model for anatomic location of infection caused by mastitis pathogens in cattle,  1923 20-7 Scale used in rating udder edema,  1989 20-8 Diagnosis of free electricity problems based on voltage difference between two points that can be accessed by an animal with resultant current flow,  1991 21-1 Causes and diagnostic features of recumbency of more than 8 hours in duration in adult horses,  2004 21-2 Species of streptococci isolated from the pig with principal locations,  2045 21-3 Differentiation of acute vesicular disease,  2064 21-4 Test methods available for the diagnosis of equine viral arteritis and their purpose,  2090 21-5 Definitive and intermediate hosts for Sarcocystis spp.–associated infections in agricultural animals,  2138 21-6 Summary of the theilerioses of domestic ruminants,  2144 21-7 Summary of the trypanosomoses of domestic animals and humans,  2150 21-8 Conditions associated with secondary copper deficiency,  2163 21-9 Copper levels of soils and plants in primary and secondary copper deficiency,  2166 21-10 Concentrations of copper in plasma, liver, milk, and hair; dietary intake and ratios of copper and its antagonists in normal, marginal, and copper-deficient situations,  2170 21-11 Venomous snakes of importance: taxonomy, geographic range, and major venom effects (Prepared by Daniel E Keyler, Pharm. D., FAACT),  2177 21-12 Plants causing glucosinolate poisoning,  2202

VetBooks.ir

List of Illustrations 1-1 1-2

1-3

1-4

2-1 4-1

4-2

Making a diagnosis.,  3 Effect of prevalence on positive and negative predictive values of a test with set sensitivity and specificity. The prevalence in this example is prevalence of failure to recover by recumbent cows (i.e., to stand), and the curves represent the consequent NPV and PPV for a serum aspartate aminotransferase activity of 171 U/L. (Reproduced with permission from Shpigel et al. Vet Rec 2003; 152:773.),  25 A decision tree for choosing between two interventions. (With permission from Fetrow J et al. J Am Vet Med Assoc 1985; 186:792-797.),  27 Example of the construction and use of a decision tree. The sources of probabilities and dollar values are discussed in the text. (a) The skeleton of the decision tree with a decision [treat (Tx) versus do not treat) and chance outcomes [recovery (REC) or spontaneous recovery (SPREC) versus continued cyst (CYST)]. (b) Probabilities and previously calculated outcome values are placed on the tree. (c) Expected costs of decision alternatives have been calculated and written in balloons above the chance nodes. (d) At this decision node, the correct choice is no treatment because it is cheaper ($72.96 versus $78.12). Double bars mark the pathway that is not chosen (treatment). The value $72.96 is then the outcome cost for this decision node. The value is used in the calculation of the best alternative to the previous decision node, because the process is repeated from right to left (not shown). (With permission from White ME, Erb HN. Comp Cont Educ Pract Vet 1982; 4:S426-S430.),  28 Examination of the herd with the objective of making a diagnosis.,  31 Relationship between environmental temperature, heat production rate, and body core temperature in agricultural animals. LCT, lower critical temperature; UCT, upper critical temperature. (Adapted from Kadzere et al. Livestock Prod Sci, 2002; 77:59-91.),  46 The nociceptive pathway in cattle, identifying the anatomic location of analgesic drug activity. Effective analgesia is best achieved using a multimodal approach that incorporates the administration of two or more pharmaceutical agents that attenuate or abolish the transmission,

4-3

4-4

4-5

5-1 5-2

5-3

5-4 5-5 5-6

modulation, and perception of pain. (Reproduced with permission from Coetzee JF. Vet Clin North Am Food Anim Pract 2013;29:13.),  81 Summary of system to estimate body condition scoring (BCS) in dairy cattle, ranging in score from 1.0 (could not be skinnier) to 5.0 (could not be fatter).,  91 Body condition scoring system modified from earlier work. (From Isensee A et al. Animal 2014; 8(12):1971-1977.),  92 Location of the site for ultrasonographic measurement of backfat thickness in dairy cattle from the left lateral and dorsal views (top two panels) and ultrasound image from an overconditioned cow with a back fat thickness of 34 mm. (Reproduced with permission from Schroder UJ, Staufenbiel R. J Dairy Sci 2006;89:1-4.),  93 Etiology and pathogenesis of dehydration.,  114 A, Association between eye recession into the orbit and dehydration as a percent of body weight in milk-fed calves with experimentally induced diarrhea and dehydration. The filled circles in the left panel are individual data points, the solid line is the linear regression line, and the dashed lines are the 95% confidence interval for prediction. Intravenous fluid is recommended when dehydration is estimated at 8% or more of body weight, equivalent to an eye recession into the orbit of 4 mm or more. B, The calf has an 8-mm eye recession into the orbit, equivalent to being 14% dehydrated. (Reprinted with permission from Constable PD et al. J Am Vet Med Assoc 1998;212(7):991996.),  115 Hemoglobinuria in a Holstein–Friesian heifer calf that had not been provided free access to water. The calf voluntarily drank 5 L in 5 minutes and voided red-tinged urine (on floor and in white container) 30 minutes later.,  116 Etiology and pathogenesis of hyponatremia.,  117 Types of dehydration.,  117 Calf with neurologic signs of hypernatremia, including abnormal mentation and posture and fasciculation of facial muscles. (From Byers SR, Lear AS, Van Metre DC: Sodium balance and the dysnatremias, Vet Clin Food Anim 2014;30:333350).,  118

5-7

Etiology and pathogenesis of hypochloremia.,  118 5-8 Etiology and pathogenesis of hypokalemia.,  119 5-9 Spider plot revealing the association between changes in two variables of the Henderson–Hasselbalch equation, plasma bicarbonate concentration (cHCO3−) and carbon dioxide tension (Pco2), on venous blood pH in 231 sick calves, most of which had diarrhea. The spider plot was obtained by systematically varying one input variable (cHCO3− or Pco2) while holding the remaining input variables at their reference values for calf venous plasma. Reference values for the two input variables for calf plasma were 29.5 mmol/L for cHCO3− (large open circles) and 53 mm Hg for Pco2 (open squares). The solid vertical and horizontal lines indicate that venous blood pH = 7.38 when cHCO3− and Pco2 are at their reference values. Note that the individual data points are displaced from the predicted pH–cHCO3− relationship. This displacement indicates that changes in plasma cHCO3− do not account for all of the changes in blood pH in sick calves. (Reproduced with permission from Constable PD, Vet Clin North Am Food Anim Pract 2014;30:295-316.),  124 5-10 Evaluation of acid-base balance using the traditional Henderson–Hasselbalch equation (A) and strong ion difference (SID) theory (B). The Henderson– Hasselbalch equation posits that blood pH is dependent on the respiratory system, as assessed by the partial pressure of carbon dioxide (Pco2), and metabolism, as assessed by the bicarbonate concentration (cHCO3−) or base excess. A, It highlights one of the fundamental flaws with using the Henderson–Hasselbalch equation in that blood pH cannot be dependent on cHCO3− because bicarbonate concentration is calculated from blood pH and Pco2. B, For comparison, this conveys that the strong ion approach to acid-base balance posits that blood pH is dependent on the respiratory system, assessed by Pco2, and on metabolism, assessed by the SID and concentration of nonvolatile buffers (Atot, such as albumin, globulin, and phosphate) in plasma. (Reproduced with permission from Constable PD: Clinical assessment of acid-base status: Strong ion difference theory, Vet Clin North Am Food Anim Pract 1999;15:447-71).,  125 xxvii

xxviii

List of Illustrations

VetBooks.ir

5-11 Spider plot revealing the association among changes in three independent variables of the simplified strong ion equation, strong ion difference (SID, open circles), carbon dioxide tension (Pco2, open squares), and the plasma concentration of nonvolatile buffers (Atot, open triangles), on venous blood pH in 231 sick calves, most of which had diarrhea. The spider plot was obtained by systematically varying one input variable (SID, Pco2, or Atot) while holding the remaining input variables at their reference values for calf venous plasma (42 mEq/L for SID), 53 mm Hg for Pco2, and 18.5 mmol/L for Atot. The solid vertical and horizontal lines indicate that venous blood pH = 7.38 when SID, Pco2, and Atot are at their reference values. Note that the individual data points are located more centrally around the predicted pH–SID relationship than for the pH–cHCO3− relationship identified in Figure 5.9. This is because changes in plasma protein concentration (and therefore Atot) caused by changes in hydration status account for some of the change in blood pH. The plot also indicates the six primary acid-base disturbances (respiratory, strong ion, or nonvolatile buffer ion acidosis and alkalosis) and the relative effect of each disturbance on blood pH in the neonatal calf. Note that changes in SID have the greatest relative effect on blood pH. (Adapted from Constable PD, Stämpfli HR, Navetat H, et al.: Use of a quantitative strong ion approach to determine the mechanism for acid-base abnormalities in sick calves with or without diarrhea. J Vet Intern Med 2005;19:581-9. IN Constable PD: Acid-Base Assessment When and How to Apply the Henderson-Hasselbalch Equation and Strong Ion Different theory, Vet Clin Food Anim. 2014;30:295-316.),  126 5-12 Etiology and pathogenesis of acidemia.,  127 5-13 Etiology and pathogenesis of alkalemia.,  129 5-14 The interrelationships among the changes in body water, electrolytes, and acid-base balance that can occur in diarrhea.,  131 5-15 Relationship between venous blood pH and anion gap (AG) in 806 neonatal calves with diarrhea. The thick line represents the result of nonlinear regression analysis: pH = log10 (39.7 − AG) + 5.92. (Reproduced with permission from Trefz FM, Constable PD, Lorenz I. J Vet Intern Med 2015;29:678-687.),  135 5-16 Relationship between venous blood pH and strong ion gap (SIG) in 806 neonatal calves with diarrhea. The

5-17

5-18

5-19

5-20

thick line represents the result of nonlinear regression analysis: pH = log10 (SIG + 32.4) + 5.84. (Reproduced with permission from Trefz FM, Constable PD, Lorenz I. J Vet Intern Med 2015;29:678-687.),  136 Optimized decision tree for treating neonatal calves with diarrhea in a field setting. Examination of the ability to stand is evaluated by lifting recumbent animals. Enophthalmos reflects a visible gap of 3 to 4 mm between the corneal surface of the eye and the caruncula lacrimalis or normal position of the lower eyelid. (From Trefz FM et al. BMC Vet Res 2012;8:238.23).,  144 Securing a 14-gauge 14-cm catheter into the jugular vein of a cow. The site of venipuncture is clipped and scrubbed for aseptic placement of a catheter. A 1-mL bleb of 2% lidocaine is placed intradermally at the proposed site of catheter insertion and a 5-mm long stab incision made through the skin, including the dermis. A, The catheter is then placed into the lumen of the vein and carefully advanced until the hub of the catheter is level with the skin. The catheter is secured by placing sutures through the skin using an 18-gauge needle and a synthetic multifilament suture material. The suture does a loop around the extension tubing near where it attaches to the hub of the catheter so that the catheter cannot back out. B, The 18-gauge needle is then passed through the ventral skin fold adjacent to the catheter, and the suture tightened to create a tunnel. C, Additional sutures are placed through the upper and lower skin folds to lengthen the tunnel and prevent excessive movement at the junction of the catheter with the hub.,  146 Administering large-volume isotonic crystalloid solutions to Holstein–Friesian cows by the jugular vein (A) and auricular vein (B).,  147 Placement of a 22-gauge 2.5-cm over the stylet butterfly catheter into the auricular vein of a calf. The ear is clipped and scrubbed for aseptic placement of a catheter, and a tourniquet is placed at the base of the ear to facilitate visualization of the auricular veins (A). The catheter is then placed into the lumen of the vein and carefully advanced (B). The tourniquet is removed and the catheter is secured to pinna by placing a 20-gauge needle though the pinna and butterfly section and tying, taking care not to distort the ear (C). Intravenous fluids are then attached and the ear bandaged, taking care not to bandage below the end of the

6-1

6-2

7-1

7-2

7-3

7-4

catheter (D). (Pictures generously provided by Dr. Joachim Berchtold, Germany.),  148 Diagram of the approximate locations of the major arteries of the posterior ear and the recommended needle insertion locations. Administration of EXCEDE Sterile Suspension into ear arteries is likely to be fatal. EXCEDE can also be administered SC at the base of the ear in a rostral direction toward the eye on the same side of the head as the ear, or administered SC at the base of the ear in a ventral direction. (Courtesy of Zoetis, Inc., https://www.zoetisus.com/products/ pages/excede_beef/TechnicalResources. aspx EXCEDE® (Cefiofur Crystalline Free Acid) Sterile Suspension.),  160 Relationship between minimum bactericidal concentration (MBC) and minimum inhibitory concentration (MIC) for bacteriostatic and bactericidal agents.,  165 A, Ultrasonographic image obtained at site 1: an abnormal amount of anechoic fluid is visible. B, Ultrasonographic image showing nonturgid fluid-filled small-intestinal loops. C, Ultrasonographic image showing turgid small-intestinal loops without wall thickening in a horse with small-intestinal obstruction. D, Ultrasonographic image showing turgid small-intestinal loops with marked wall thickening in a horse with strangulated small-intestinal obstruction.,  229 A, Left lateral view of abdomen of a normal horse. B, Left dorsal displacement of the left colon, left lateral view. The left ventral and dorsal colon is displaced lateral and dorsal to the spleen and occupies the renosplenic space. 1, liver; 2, stomach; 3, left dorsal colon; 4, left ventral colon; 5, spleen; 6, left kidney and renosplenic ligament; 7, pelvic flexure. (With permission from Johnston JK, Freeman DE. Vet Clin North Am Equine Pract 1997;13:317.),  260 Right dorsal displacement of the colon, right lateral view. The colon has passed lateral to the cecum, the pelvic flexure is displaced cranially, and the sternal and diaphragmatic flexures are displaced caudally. 1, right dorsal colon; 2, base of cecum; 3, right ventral colon; 4, liver; 5, cecum; 6, left ventral colon; 7, pelvic flexure. (With permission from Johnston JK, Freeman DE. Vet Clin North Am Equine Pract 1997;13:317.),  261 A 360 clockwise volvulus of the colon viewed from the right side. The volvulus has occurred in the direction of the arrow. 1, cecum; 2, right dorsal colon; 3, right ventral colon. (With

List of Illustrations

VetBooks.ir

7-5

7-6

7-7

7-8

8-1

8-2

8-3

permission from Johnston JK, Freeman DE. Vet Clin North Am Equine Pract 1997;13:317.),  261 Steps in correction of left dorsal displacement of the colon (renosplenic entrapment). A, Caudal view of abdomen of horse with left dorsal displacement of the colon. Entrapped colon is shown in black. K, left kidney; S, spleen. B, Injection of phenylephrine and contraction of spleen. C, Horse anesthetized and placed in right lateral recumbency. D–H, Horse rolled through dorsal recumbency to left lateral recumbency. Entrapped colon moves ventrally and then medially to the contracted spleen. (Modified with permission from Kalsbeek HC. Equine Vet J 1989;21:442.),  263 Enterolith in the large colon of a horse with colic. The scale marker is 10 cm. (Reproduced with permission from Kelleher ME, et al. JAVMA. 2014;245:126.4),  266 Lateral abdominal radiograph of a Miniature horse with severe sand accumulation. (Reproduced with permission from Hart KA, et al. Equine Vet J. 2013;45:465.1),  267 Percutaneous ultrasonographic image of an 8-month-old filly with enteritis caused by Lawsonia intracellularis and demonstrating markedly thickened small-intestinal wall (9 mm; normal 18 hours Prevention: Check colostrum after birth. Concentration specific gravity at foaling. should be > 800 mg/dL (8 g/L). Ensure foal nurses mare within 3 hours of birth. Supplement with banked colostrum. Routinely measure foal serum IgG at 18–24 hours of age.

Severe combined Failure of V(D)J immunodeficiency recombination of Arabians secondary to a defect of the catalytic subunit of DNA-protein kinase coded for by the DNA-PKcs gene.

Epidemiology: Restricted to Arabians. Autosomal inheritance Clinical signs: Adenoviral pneumonia or diarrhea develop after ~4 weeks of age.

Severe lymphopenia ( 2 months of age serum. No B lymphocytes (corresponds with declining detectable in blood. Normal colostral immunity). concentrations of T lymphocytes.

Treatment: None specific. Supportive and symptomatic, but all affected foals die. Prevention: None.

Epidemiology: Adult horses. Sporadic. Either sex. Clinical signs: Chronic or recurrent infections unresponsive to medical treatment. Meningitis. Liver disease can occur in combination with common variable immunodeficiency.

Treatment: None specific. Affected animals die because of the opportunistic infections. Prevention: Prolonged antimicrobial administration of affected horses.

Common variable Unknown in horses. immunodeficiency

Low to undetectable concentrations of IgG, IgG(T), IgM, and IgA in serum. No or few B lymphocytes in blood or lymph nodes. Elevations in serum markers of liver disease.

Treatment and prevention

Amyloidoses

VetBooks.ir

• Agammaglobulinemia of Standardbred and Thoroughbred horses, probably the result of an inherited failure to produce B lymphocytes; these horses live much longer than those affected with CID. • Selective deficiencies of one or more globulins—a deficiency of IgM in Arabian horses and Quarter horses is listed. IgM and IgA combined deficiencies with diminished but discernible levels of IgG are observed occasionally in horses. A transient hypogammaglobulinemia (absence of IgG) has been reported in one Arabian foal, which was immunodeficient until it was 3 months old and then became normal. • Selective IgG2 deficiency in Red Danish cattle • A syndrome of immunodeficiency in Fell ponies and Dale ponies2,3 • Common variable immunodeficiency is described in adult horses.4,5 • Lethal trait A46 (inherited parakeratosis) of cattle is a primary immunodeficiency influencing T lymphocytes, with impairment of cellular immunity. • Selective IgG2 deficiency of cattle causes increased susceptibility to gangrenous mastitis and other infections. It is a primary deficiency of IgG2 synthesis and is recorded in the Red Danish milk breed. • Sheep and pigs—there are as yet no recognized primary immunodeficiencies in these species.

SECONDARY IMMUNE DEFICIENCIES Secondary immune deficiencies include the following: • Failure of transfer of passive immunity (i.e., of antibodies from colostrum to the offspring) is well known as the commonest cause of deficient immunity in the newborn and is discussed in Chapter 20. • Atrophy of lymphoid tissue and resulting lymphopenia are associated with the following: • Viral infections such as equine herpesvirus in newborn foals, rinderpest, bovine virus diarrhea, swine fever, porcine circovirus, and hog cholera. All of these cause lymphatic tissue suppression and a diminished immunoresponsiveness. • Bacterial infections such as Mycoplasma spp. and Mycobacterium paratuberculosis have approximately the same effect as the viral infections. • Physiologic stress, such as birth, may cause immunosuppression in the fetus, making it very susceptible to infection in the period immediately after birth. There is a similar depression of

immunologic efficiency in the dam immediately after parturition, which, for example, leads to periparturient rise of worm infestation in ewes. • Toxins such as bracken, tetrachloroethylene-extracted soybean meal, T2 mycotoxin, and atomic irradiation suppress leukopoiesis. Immunosuppression is also attributed to many environmental pollutants, including polychlorinated biphenyls, 2,4,5-T contaminants, DDT, aflatoxin, and the heavy metals. • General suppression of immune system responsiveness; examples include the following: • Glucocorticoids administered in large doses or over long periods reduce the activity of neutrophils and the number of circulating lymphocytes, although the reduction varies widely between species. The production of antibodies is also reduced. • Nutritional deficiency, especially of zinc, pantothenic acid, calcium, and vitamin E, causes general suppression. A total caloric deficiency has a similar effect. Addition of certain trace elements, such as copper, iron, zinc, and selenium, in animal feeds is necessary for an adequate immunity. Selenium, alone or in combination with vitamin E, can enhance antibody responses, whereas its deficiency results in immunosuppression. Selenium supplementation in animal feeds is important to enhance both antibody production and phagocytic activity of neutrophils. In cattle, copper deficiency induced by molybdenum or iron can cause an impairment in the ability of neutrophils to kill ingested Candida albicans. Nutrients that stimulate disease resistance when administered to animals deficient in these nutrients include carotenoids; vitamins A, E, and C; zinc; manganese; copper; and selenium. Neonatal calves may have low reserves of carotene and vitamins A and E and are dependent on obtaining them from colostrum, which contains highly variable quantities. Administration of drugs that impair folate metabolism can induce anemia and depletion of white blood cells, with subsequent bacterial infection. • Experimentally, a protein–energy malnutrition in neonatal calves results in loss of body weight and decreased lymphocyte interleukin-2 activity and lymphocyte proliferation compared with calves of similar age.

755

• Exposure to cold and heat stress for periods of several weeks in duration • Events associated with parturition, in particular glucocorticoid release, that impair innate immunity • Alloimmune disease that suppresses bone-marrow activity6 REFERENCES

1. Takasu M, et al. J Vet Med Sci. 2008;70:1173. 2. Fox-Clipsham LY, et al. PLoS Genet. 2011;7. 3. Fox-Clipsham L, et al. Vet Rec. 2009;165:289. 4. Tallmadge RL, et al. Mol Immunol. 2012;51:169. 5. Tallmadge RL, et al. J Clin Immunol. 2012;32:370. 6. Euler KN, et al. BMC Vet Res. 2013;9.

Amyloidoses The amyloidoses are a group of diseases characterized by the deposition of an extracellular proteinaceous substance, amyloid, in the tissues, with subsequent disruption of normal tissue architecture that eventually leads to organ dysfunction. Amyloidosis in farm animals usually occurs in association with a chronic suppurative process elsewhere in the body and is the result of accumulation of AA amyloid. Another form of the disease involves accumulation of AL amyloid, especially as localized disease in horses.

ETIOLOGY AND EPIDEMIOLOGY

Amyloidosis occurs rarely, and when it does occur it is most common in animals exposed systemically and repeatedly to antigenic substances. Examples include repeated injections of antigenic material for commercial production of hyperimmune serum and long-standing suppurative diseases or recurrent infection, as in Chédiak–Higashi syndrome. Severe strongylid parasitism in the horse has been reported as a cause. Holstein calves with bovine leukocyte adhesion deficiency have accumulation of amyloid in tissue, although this is not the primary disease. Many cases of amyloidosis in large animals are without apparent cause. The incidence of visceral AA amyloidosis in slaughtered cattle in a group of 302 cattle older than 4 years of age in Japan was 5.0%; rates previously reported from Japan and other countries ranged from 0.4% to 2.7%. Systemic AA amyloidosis associated with tuberculosis has been described in a European wild boar. Systemic amyloidosis occurs in goat kids with chronic arthritis associated with seroconversion to Erysipelothrix rhusiopathiae. AA renal amyloidosis in cattle is often associated with traumatic reticuloperitonitis, metritis, mastitis, or pododermatitis, although 5 of 25 cows with AA amyloidosis did not have coexisting chronic inflammatory disease.1 Out of 16,000 horses referred for clinical examination to a veterinary teaching hospital over a period of 13 years, 9 horses were identified as having amyloidosis. Cutaneous

756

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

amyloidosis has been associated with malignant histiocytic lymphoma in horses. A case of cardiac amyloidosis causing heart failure in a 16-year-old Thoroughbred gelding has been described. The disease resulted from accumulation of AL amyloid. The AL form of amyloidosis is characteristically associated with unstable monoclonal immunoglobulin light chains produced by plasma-cell dyscrasia and resulting in deposition of AL fibrils.

PATHOGENESIS

How amyloid is formed is uncertain, but hyperglobulinemia is commonly present. This fact, together with the circumstances under which it occurs, suggests an abnormality of the antigen–antibody reaction. Amyloidoses are classified by the types of amyloid protein deposited. AA amyloid is derived from serum amyloid-A protein (SAA), which is an acute-phase reactant produced by hepatocytes. However, increased concentrations of SAA alone are not sufficient to cause amyloidosis. AA (secondary) amyloidosis is associated with recurrent acute or chronic infections, inflammatory disease, or neoplasia. Extensive amyloid deposits may occur in the spleen, liver, or kidneys and cause major enlargement of these organs and serious depression of their functions. The commonest form that is clinically recognizable in animals is renal amyloidosis. This presents as a nephrotic syndrome with massive proteinuria and a consequent hypoproteinemia and edema. Terminally, the animal is uremic, becoming comatose and recumbent. The edema of the gut wall and its infiltration with amyloid create the conditions necessary for the development of diarrhea. In horses, cases of multiple cutaneous lesions are recorded. The amyloid is present in 5- to 25-mm diameter nodes in the skin of the head, neck, and pectoral regions. Rare cases of involvement of the upper respiratory tract (nasal cavities, pharynx, larynx, guttural pouch and lymph nodes of the head and neck, and conjunctiva) occur in horses.2 The amyloid material deposited in these tissues is usually of the AL form,2 whereas systemic disease is almost always the AA form. AL amyloidosis is also reported in an adult cow with bovine leukocyte adhesion deficiency.

CLINICAL FINDINGS

Many cases of amyloidosis are detected incidentally at necropsy. The cutaneous form in horses is characterized by the presence of hard, nonpainful, chronic plaques in the skin. Most of the lesions, which can be widespread and severe, are on the sides of the neck, shoulders, and head. Respiratory tract involvement in the horse is usually limited to the nasal cavities,2 and this may cause dyspnea. There is deposition of AA amyloid

Fig. 11-3  Lactating Brown Swiss cow with massive persistent proteinuria and severe hypoalbuminemia (serum albumin concentration, 0.7 g/dL) as a result of advanced renal amyloidosis. Note the ventral abdominal and submandibular edema secondary to the marked hypoalbuminemia. An enlarged left kidney was detected per rectal palpation, and renal biopsy confirmed the diagnosis of renal amyloidosis.

in the ciliary body of horses with recurrent uveitis, although the clinical importance of this finding is uncertain.3 Chronic heart failure as a result of cardiac amyloidosis secondary to systemic amyloidosis in a 16-year-old gelding was characterized clinically by weight loss, dysphagia, recurrent episodes of esophageal obstruction, and anorexia of a few weeks in duration. Ventral edema, tachycardia, and irregular heart rate associated with atrial fibrillation were present. The clinical findings were consistent with biventricular heart failure from ventricular dysfunction, atrial fibrillation, and pulmonary hypertension. The amyloid was of the AL form. A case of systemic AL amyloidosis associated with multiple myeloma in a horse was characterized clinically by rapid weight loss, muscle atrophy, soft unformed feces, and ventral edema. Amyloidosis in this situation is considered a paraneoplastic disease.4 Hemoperitoneum and acute death secondary to splenic or hepatic rupture occurs in horses with systemic amyloidosis. Clinical cases in cattle are usually secondary to traumatic reticuloperitonitis, mastitis, metritis, or pododermatitis and are characterized by emaciation and enlargement of the spleen, liver, or kidneys; involvement of the kidney causes proteinuria and is often accompanied by profuse and chronic diarrhea, polydipsia, and anasarca.1 In cattle the grossly enlarged left kidney is usually palpable per rectum. Cases can manifest within 2 weeks of calving. They are characterized by anorexia, watery diarrhea, anasarca, rapid emaciation, and death in 2 to 5 weeks. Corpora amylacea are small, round concretions of amyloid material found in the

mammary tissue of cows. They are usually inert but may cause blockage of the teat canal.

CLINICAL PATHOLOGY

An extreme and persistent proteinuria should suggest the presence of renal amyloidosis. Electrophoretic studies of serum may be of value in determining the presence of hyperglobulinemia. Serum alpha-globulin concentration is usually elevated and albumin concentration markedly depressed. Hypoproteinemia can be marked, with ventral edema frequently being present when the serum albumin concentration is less than 1.0 g/dL (Fig. 11-3). Horses with hepatic amyloidosis have elevated activities of gamma-glutamyltransferase and, to a lesser extent, bile acids. In cattle there is hypoproteinemia, hypoalbuminemia, hypocalcemia, hyperfibrinogenemia, hypomagnesemia, high serum urea and creatinine concentration, and low-specific-gravity urine.1 Serum amyloid A and haptoglobin concentrations in the serum of cows with amyloids are higher than those in healthy cows, but not compared with cows with chronic inflammatory disease in the absence of amyloidosis.5 Biopsy of cutaneous plaques is an accurate diagnostic technique.

NECROPSY FINDINGS

Amyloid can be detected in most organs of cows with systemic AA amyloidosis by histologic examination, including the liver, kidney, thyroid gland, adrenal gland, gastrointestinal mucosa, heart, lung, lymph nodes, ovary, hypophysis, uterus, mammary gland, and skeletal muscle.6,7 Grossly affected organs are enlarged and have a pale, waxy

Amyloidoses

VetBooks.ir

appearance. In the spleen, the deposits are circumscribed, whereas in the liver and kidneys they are diffuse. In a horse with systemic AL amyloidosis associated with multiple myeloma, diffuse gastrointestinal hemorrhage, thickened jejunal mucosa, and splenomegaly were present. The pathology of AA amyloidosis in domestic sheep and goats has been described. Most sheep with amyloidosis had pneumonia and other sites of chronic inflammation. Amyloid was detected in all grossly affected kidneys using Congo Red staining. Experimental induction of amyloidosis secondary to pneumonia in sheep results in deposition of amyloid in the gastrointestinal tract from the tongue to the rectum, with the most abundant deposition in the duodenum.8 Other body systems have only mild deposition of amyloid.8 Deposits of amyloid in tissues may be made visible by staining with aqueous iodine. Amyloid is detected as green birefringence of Congo Red–stained tissues viewed under polarized light. AA and AL amyloidosis can be differentiated by treatment of tissue sections with potassium permanganate. Tissue containing AA will lose its green birefringence after treatment with potassium permanganate, whereas tissue containing AL will continue to appear green after Congo Red staining and viewing under polarized light. The Shtrasburg method is now available for the identification of AA amyloid and to distinguish it from amyloid types in a large number of domestic and wild animals. DIFFERENTIAL DIAGNOSIS Enlargement of parenchymatous organs associated with chronic suppurative processes should arouse suspicion of amyloidosis, especially if there is emaciation and marked proteinuria. Pyelonephritis, nonspecific nephritis, and nephrosis bear a clinical similarity to amyloidosis.

TREATMENT There is no effective treatment of the systemic disease. The localized disease as occurs in the upper respiratory tract of horses can be treated by surgical excision, but the results are not encouraging.

ZOONOTIC POTENTIAL

There is discussion of the potential for AA amyloidosis to be a transmissible disease, although there is no evidence of a risk to human health from ingestion of meat and organs from cows with clinically inapparent amyloidosis.9 REFERENCES

1. Elitok OM, et al. J Vet Int Med. 2008;22:450. 2. Ostevik L, et al. Acta Vet Scand. 2014;56. 3. Ostevik L, et al. J Comp Pathol. 2014;151:228.

4. Axiak S, et al. Equine Vet Educ. 2012;24:367. 5. Takahashi E, et al. J Vet Med Sci. 2007;69:321. 6. Yamada M, et al. J Vet Med Sci. 2006;68:725. 7. Murakami T, et al. Amyloid. 2012;19:15. 8. Biescas E, et al. J Comp Pathol. 2009;140:238. 9. Murakami T, et al. Vet Pathol. 2014;51:363.

ALLERGY AND ANAPHYLAXIS Immune-mediated diseases in animals included here are those in which the fundamental abnormality is an exaggerated or misdirected immune response. Many diseases, and in particular infectious diseases, have an important component of their pathophysiology that is attributable to immune responses to the inciting agent. Although these responses can sometimes be deleterious to the animal, they are part of an expected immunologic reaction to the infectious agent. Examples of where a component of the immune response to an infection has an important role in the pathophysiology of the clinical disease include pneumonia in calves infected by Mycoplasma bovis, pneumonia in foals secondary to equine influenza virus infection, and enteritis and colitis secondary to salmonella infection. Diseases manifest by allergy or anaphylaxis are characterized by exaggerated immune responses or reactions to otherwise innocuous stimuli. Examples in large animal medicine include immediate hypersensitivity reactions (anaphylaxis), milk allergy in Jersey cattle, dermatologic diseases (such as Queensland itch), neonatal isoimmune erythrolysis of foals, and purpura hemorrhagica. There are four major mechanisms for the induction of a hypersensitivity response. They are classified as types I through IV based on the immune mechanism that elicits the disease state. Types I through III are antibody-mediated responses to antigen and include such conditions as systemic anaphylactic shock (type I), autoimmune hemolytic anemia (type II), and the local Arthus reaction (type III). Type IV hypersensitivity is caused by the induction of sensitized T lymphocytes and thus has a cell-mediated mechanism. The following description is minimalistic, including minimal mention of Th1 and Th2 responses, and readers are referred to texts dealing with veterinary immunology for more detail.

TYPE I

Type I disease is caused by binding of specific antigens by antibodies with local and/or systemic responses that occur within minutes. The antibody involved is almost always IgE, or its functional equivalent in species in which this role is fulfilled by a different class of antibody, that is bound to specific receptors on the surface of mast cells. Binding of the antigen to the surface-bound IgE precipitates release of inflammatory and vasoactive mediators, including histamine, various prostaglandins and leukotrienes, and

757

cytokines (IL-4, IL-5, IL-6, IL-13, tumor necrosis factor alpha, and others), from the cells (“degranulation”), with subsequent local reactions of hyperemia and heat and systemic reactions such as constriction of smooth muscles (importantly, bronchial smooth muscle). Degranulation of mast cells also attracts eosinophils to the site with their subsequent degranulation and release of a wide variety of compounds, including cytokines, chemokines, enzymes, oxidants, prostanoids, and cationic granule proteins. The difference in manifestation of acute, immediate-type hypersensitivity reactions between species appears to depend largely on differences in the tissue site of antibody binding and the route of entry into the body of the inciting allergen. For example, insect bite hypersensitivity (Queensland itch) is the result of an IgE-mediated response to antigens in the saliva of biting insects—the inciting allergen is deposited in the skin when the insect bites the host. The disease is only manifest in horses that do not have an appropriate tempering immunologic reaction.1 The high incidence of atopic hypersensitivity with familial predisposition seen in humans and dogs does not occur as frequently in large animals, although it is notable that there are breed differences in cell-mediated and antibody-mediated hypersensitivity reactions in cattle.2,3 Type I hypersensitivity is considered to be the mechanism for selfcure of gastrointestinal parasitism in sheep. Anaphylaxis is the systemic manifestation of widespread and massive release of inflammatory mediators from mast cells. Signs are acute, occurring within minutes of exposure to the allergen, and severe, resulting in bronchoconstriction and leading to death in many cases. Treatment involves prevention of exposure or removal of inciting allergens, administration of corticosteroids, and, in anaphylaxis, administration of drugs that counteract the severe bronchoconstriction (epinephrine). Antihistamines are minimally effective, likely because histamine is only one of numerous inflammatory activators released by mast cells and eosinophils.

TYPE II

Type II hypersensitivity reaction is caused by the binding of antibodies to antigenic sites on specific cell types (antibody-mediated cell cytotoxicity). The disease can occur because of the presence of antibodies against specific natural proteins on cell surfaces, such as occurs with neonatal isoerythrolysis or reactions to blood transfusions, or when foreign antigens, such as viruses or virus particles, bind to cell surfaces. Diseases include neonatal isoimmune hemolysis and thrombocytopenia.

TYPE III

Type III hypersensitivity reaction occurs as a result of formation of antibody–antigen

758

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

complexes, which then induce an inflammatory response in the host tissue. Examples include Arthus-type reaction or the Arthus phenomenon, which is evident as induration, erythema, edema, hemorrhage, and necrosis of the skin a few hours after intradermal injection of antigen into a previously sensitized animal; deposition of antibody– antigen complexes in the glomeruli, with resultant inflammation and tissue damage evident as glomerulonephritis; purpura hemorrhagica in equids; and in many viral diseases in which viruses are not neutralized by circulating antibodies (equine infectious anemia). The lesion results from the precipitation of antigen–antibody complexes, which causes complement activation and the release of complement fragments that are chemotactic for neutrophils; large numbers of neutrophils infiltrate the site and cause tissue destruction by release of lysosomal enzymes.

TYPE IV

Cell-mediated or delayed hypersensitivity is the basis for tuberculin and bovine paratuberculosis skin tests in cattle. The response is mediated by T cells and natural-killer (NK) cells. Allergic contact dermatitis, a cutaneous from of type IV hypersensitivity, is commonly identified in dogs and cats, but less so in large animals. Cattle can develop an allergic reaction to calcium cyanamide, a nitrogenous fertilizer, evident as allergic contact dermatitis.4 The disease occurred in 9 of 250 dairy cattle housed on flooring to which calcium cyanamide had been added to reduce the risk of environmental mastitis. The cows developed alopecia, erythema, and crusting and pruritus of the udder, teats, ventral abdomen, and dewlap—all areas that contacted the ground surface. The disease can be severe and markedly affect the health and well-being of affected cows. Diagnosis is confirmed by skin-patch testing using cyanamide. 4 Cattle can also develop delayed hypersensitivity to rubber in milking machines.5 A more severe form of type IV–mediated skin disease is toxic epidermal necrolysis (also known as Stevens–Johnson syndrome). This disease in humans is often associated with administration of medications. It occurs in calves infected with Mycoplasma bovis and is evident as pneumonia, arthritis, and severe skin lesions manifest as marked thickening of the epidermis, detachment of the epidermis from the dermis, and blisters in the detachment sites.6 Affected calves recovered, apparently in response to treatment with antimicrobials, a single administration of corticosteroid and pentoxifylline. Erythema multiforme is a milder form of cutaneous expression of delayed hypersensitivity. It occurs in horses and cattle and is manifest as sudden onset of roughly symmetric erythematous wheals on the neck and dorsum with peripheral expansion and

Anaphylaxis is an acute disease of often lifethreatening severity caused by an antigen– antibody (IgE) reaction. If severe it can result in anaphylactic shock.

Hypersensitivity reactions are sometimes observed at a higher incidence than normal in certain families and herds of cattle. Anaphylactic reactions can occur in the following circumstances: • Repeated injection of biological preparations • Repeated blood transfusions from the same incompatible donor or donors9 • Repeated injections of vaccines (e.g., those against foot-and-mouth disease and rabies) • Injection of penicillin—although many presumed penicillin-induced anaphylactic reactions are in fact reactions to inadvertent intravenous administration of procaine or benzathine • Similar rare occurrences after the injection of lyophilized Brucella abortus strain 19 vaccine and Salmonella vaccine • Assumed anaphylactic reaction to ingested protein occurs in animals at pasture or in the feedlot. • Cows, especially Channel Island cattle, can develop anaphylaxis when milking is stopped because the cows are being dried off; severe urticaria and respiratory distress occur 18 to 24 hours later. • A systemic reaction after Hypoderma spp. larvae are killed in their subcutaneous sites might be anaphylactic, but is more likely to be a toxic effect from breakdown products of the larvae. • After inadvertent intravenous administration of mare’s milk to a foal10

ETIOLOGY

PATHOGENESIS

central clearing forming donut-like lesions. Lesions can persist for days to weeks and be moderately painful. Scaling, crusting, and alopecia are unusual.7 Lesions heal spontaneously. A similar disease is associated with cutaneous equine herpesvirus-5 infection, although the skin lesions were most severe on the muzzle and face.8 Delayed hypersensitivity reactions can contribute to the pathology of many diseases, such as mycoplasmal pneumonia in swine, but those are considered clinically under their initiating etiology.

TREATMENT

The treatment of allergic states is based on immediate treatment of signs of inflammation or allergy, usually by the administration of corticosteroids, antihistamines, or, in anaphylaxis, epinephrine, and prevention of continued exposure to the inciting allergen. Antihistamines have very limited usefulness, whereas corticosteroids have wide applicability and potent efficacy. The NSAIDs, including such drugs as flunixin meglumine, phenylbutazone, and meclofenamic acid, all inhibit prostaglandin synthesis and thus reduce inflammation but have only slight effect in treating allergic diseases.

ANAPHYLAXIS AND ANAPHYLACTIC SHOCK

Most commonly, severe anaphylactic reactions are seen in farm animals following the parenteral administration of a drug or biological product. Other routes of entry of the allergen, such as via the respiratory or gastrointestinal tract, can also result in anaphylactic reactions. The reaction can occur at the site of exposure or in other areas. The disease occurs because the animal is sensitized to the inciting allergen by previous exposure. The initial exposure usually does not result in any immediate clinical abnormalities, but subsequent exposure of the animal to the antigen results in rapid degranulation of mast cells and, subsequently, eosinophils, with widespread release of vasoactive and inflammatory mediators resulting in anaphylactic shock. Although severe anaphylactic reactions occur usually after a second exposure to a sensitizing agent, reactions of similar severity can occur with no known prior exposure. In large animal practice this is most likely to occur after the injection of sera and bacterins, particularly heterologous sera and bacterins in which heterologous serum has been used in the culture medium.

Anaphylactic reactions occur as the result of antigen reacting with cell-bound antibody. In humans, horses, and dogs a specific class of reaginic antibody, IgE, has been identified and has particular affinity for fixed tissue mast cells. The tissue distribution of mast cells in part accounts for the involvement of certain target organs in anaphylactic reactions in these species. Homocytotropic antibody has been detected in farm animals, but the classes of antibodies involved in anaphylactic reactions have not been fully identified and might be diverse. Anaphylactic antibodies can be transferred via colostrum. Antigen–antibody reactions occurring in contact with, or in close proximity to, fixed tissue mast cells, basophils, and neutrophil leukocytes result in the activation of these cells to release pharmacologically active substances that mediate the subsequent anaphylactic reaction. These substances include biogenic amines, such as histamine, serotonin, and catecholamines; vasoactive polypeptides, such as kinins, cationic proteins, and anaphylatoxins; vasoactive lipids, such as prostaglandins; and slow-reacting substance of anaphylaxis (SRS-A), among others. Knowledge of the type and relative

Amyloidoses

VetBooks.ir

importance of pharmacologic mediators of anaphylaxis in farm animals rests with studies of severe anaphylactic reactions that have been induced experimentally, but it is likely that these mediators are also of significance in less severe reactions. From these studies it appears that histamine is of less importance as a mediator in farm animals than in other species and that prostaglandins and SRS-A are of greater importance. Bradykinin and 5-hydroxytryptamine (5-HT) are also known to act as mediators in cattle, but the reactions in all species are complex and involve a sequence of mediator effects. In horses, there are four phases in the development of the anaphylactic response. The first is acute hypotension combined with pulmonary arterial hypertension 2 to 3 minutes after the injection of the triggering agent; it coincides with histamine release. In the second phase, blood plasma 5-HT levels rise, and central venous blood pressure rises sharply at about 3 minutes and onward. The third phase commences at about 8 to 12 minutes and is largely reflex and manifested by a sharp rise in blood pressure and alternating apnea and dyspnea. Finally, there is a second and more protracted systemic hypotension as a result of prostaglandin and SRS-A influence, which persists until the return to normality. In cattle, there is a similar diphasic systemic hypotension with marked pulmonary venous constriction and pulmonary artery hypertension. An increase in mesenteric venous pressure and mesenteric vascular resistance causes considerable pooling of blood on the venous side of the mesenteric vessels. In both cattle and horses these reactions are accompanied by severe hemoconcentration, leukopenia, thrombocytopenia, and hyperkalemia. Sheep, goats, and pigs also show a largely pulmonary reaction.11 In horses and cattle, the marked changes in vascular tone coupled with increased capillary permeability, increased secretion of mucous glands, and bronchospasm are the primary reactions leading to the development of severe pulmonary congestion, edema, and emphysema and edema of the gut wall. Less severe reactions are also dependent on the effect of mediators on capillary permeability, vascular tone, and mucous gland secretion. The major manifestation depends on the distribution of antibody-sensitized cells and of susceptible smooth muscle in the various organs. In cattle, reactions are generally referable to the respiratory tract, but the alimentary tract and skin are also target organs. Sheep and pigs show largely a pulmonary reaction, and horses manifest changes in the lungs, skin, and feet. Sensitization of an animal requires about 10 days after first exposure to the antigen and persists for a very long time, usually months or years.

CLINICAL FINDINGS Cattle In cattle, initially there is a sudden onset of severe dyspnea, muscle shivering, and anxiety. In some cases, there is profuse salivation, in others moderate bloat, and yet others diarrhea. After an incompatible blood transfusion, the first sign is often hiccough. Additional signs are urticaria, angioneurotic edema, and rhinitis (Fig. 11-4). Muscle tremor can be severe, and a rise in temperature to 40.5° C (105° F) is often be observed. On auscultation of the chest there can be increased breath sounds, crackles if edema is present, and emphysema in the later stages if dyspnea has been severe. In most surviving cases the signs have usually subsided within 24 hours, although dyspnea may persist if emphysema has occurred. In natural cases the time delay after injection of the reagin intravenously is about 15

759

to 20 minutes, but in experimentally induced cases a severe reaction may be evident within 2 minutes of the injection and death within 7 to 10 minutes. Clinical signs include collapse, dyspnea, wild paddling, nystagmus, cyanosis, cough, and the discharge of a creamy, frothy fluid from the nostrils. Recovery, if it occurs, is complete in about 2 hours. Sheep, Goats, and Pigs In sheep, goats, and pigs, acute dyspnea is common.11 Goats with disease induced by sensitization to horse serum had respiratory distress, evident as increased respiratory rate, irregular respiration, coughing, abnormal lung sounds, reluctance to move, shivering or muscle tremors, paddling, and kicking.11 Horses In the horse, naturally occurring anaphylactic shock is manifested by severe dyspnea,

A

B Fig. 11-4  A and B, Urticaria in a Holstein–Friesian cow following antibiotic administration. Note the presence of multiple small raised bumps in multiple areas of the skin and the edema of the eyelid.

760

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

distress, recumbency, and convulsions. Death can occur within less than 5 minutes, but it usually requires about an hour. Laminitis and angioneurotic edema are also common signs in the horse. Experimentally induced anaphylaxis can be fatal but not in such a short time. Within 30 minutes of injecting the reagin the horse has anxiety, tachycardia, cyanosis, and dyspnea. These signs are followed by congestion of conjunctival vessels, increased peristalsis, fluid diarrhea, generalized sweating, and erection of the hair. If recovery occurs, it is about 2 hours after the incident began. Death, if it occurs, takes place about 24 hours after the injection. Pigs In pigs, experimentally produced anaphylactic shock can be fatal within a few minutes, with systemic shock being severe within 2 minutes and death occurring in 5 to 10 minutes. The disease appears to occur in only one phase, in contrast to the four fairly distinct states in horses. Labored respiration, severe cyanosis, vomiting, and edema of the larynx, stomach, and gallbladder are the usual outcome.

CLINICAL PATHOLOGY

There are no pathognomonic changes in serum biochemistry or hematological variables. There is a marked increase in packed cell volume, a high plasma potassium concentration, and neutropenia. Tests for sensitivity to determine the specific sensitizing substance are rarely carried out for diagnostic purposes, but their use as an investigation tool is warranted. Serologic tests to determine the presence of antibodies to plant proteins in the diet have been used in this way.

NECROPSY FINDINGS

In acute anaphylaxis in young cattle and sheep the necropsy findings are confined to the lungs and are in the form of severe pulmonary edema and vascular engorgement. In adult cattle there is edema and emphysema without engorgement. In protracted anaphylaxis produced experimentally in young calves, the most prominent lesions are hyperemia and edema of the abomasum and small intestines. In pigs and sheep pulmonary emphysema is evident, and vascular engorgement of the lungs is pronounced in the latter. Pulmonary emphysema and widespread petechiation in the horse may be accompanied by massive edema and extravasations of blood in the wall of the large bowel. There may also be subcutaneous edema and lesions of laminitis. DIFFERENTIAL DIAGNOSIS A diagnosis of anaphylaxis can be made with confidence if a foreign protein substance has been injected within the preceding

hour, but should be made with reservation if the substance appears to have been ingested. Characteristic signs as described previously should arouse suspicion, and the response to treatment may be used as a test of the hypothesis. Acute pneumonia may be confused with anaphylaxis, but there is usually more toxemia, and the lung changes are more marked in the ventral aspects; in anaphylaxis there is general involvement of the lung. • Inadvertent intravenous administration of vasoactive compounds, such as procaine as procaine penicillin, can mimic signs of anaphylaxis in that animals collapse acutely. Such cases usually lack the characteristic abnormalities on postmortem examination.

TREATMENT Treatment should be administered immediately; a few minutes’ delay can result in the death of the animal. Epinephrine is the most effective treatment for anaphylaxis and anaphylactic shock. Epinephrine administered intramuscularly (or one-fifth of the dose given intravenously) is often immediately effective, with the signs abating while the injection is being made. Corticosteroids potentiate the effect of epinephrine and can be given immediately following epinephrine. Antihistamines have been considered and were used commonly in the past, but they are likely ineffective, based on studies in humans, because they antagonize only one of many inflammatory mediators involved in the disease. The identification of mediators other than histamine in anaphylactic reactions in farm animals has led to studies of the effectiveness of drugs more active against these mediators than antihistamines. Acetylsalicylic acid, sodium meclofenamate, and diethylcarbamazine have all shown ability to protect against experimentally induced anaphylaxis in cattle and horses. One of the important clinical decisions, especially in horse practice, is to decide whether an animal is sufficiently hypersensitive to be at risk when being treated. An acute anaphylactic reaction, and even death, can occur soon after intravenous injection of penicillin into a horse. In suspect cases it is customary to conduct an intradermal or a conjunctival test for hypersensitivity with a response time of about 20 minutes, but these tests have their limitations. The types of sensitivity are not necessarily related, there is no sure relationship between anaphylactic sensitivity and either skin (or conjunctival) sensitivity or circulating antibody, and the test often gives false negatives. The reason why some animals develop systemic hypersensitivity and some develop cutaneous hypersensitivity is unknown.

OTHER HYPERSENSITIVITY REACTIONS Other hypersensitivity reactions include anaphylaxis of a less severe degree than anaphylactic shock and cases of cell-mediated delayed hypersensitivity. The resulting clinical signs vary depending on the tissues involved, but are usually localized and mild.

ETIOLOGY

Exposure to any of the etiologic agents described under anaphylaxis may result in this milder form of hypersensitivity. Exposure may occur by injection, by ingestion, by inhalation, or by contact with the skin.

PATHOGENESIS

In anaphylactic reactions the clinical signs may depend on the portal of entry. Thus ingestion may lead to gastrointestinal signs of diarrhea, and inhalation may lead to conjunctivitis, rhinitis, and laryngeal and bronchial edema. Cutaneous lesions can result from introduction of the reagin via any portal. They are usually manifested by angioedema, urticaria, or a maculopapular reaction. All the lesions result from the liberation of histamine, serotonin (5-HT), and plasma kinins, as in anaphylactic shock.

CLINICAL FINDINGS

In ruminants, inhalation of a sensitizing antigen can cause the development of allergic rhinitis. On ingestion of the sensitizing agent there may be a sharp attack of diarrhea and the appearance of urticaria or angioneurotic edema; in ruminants mild bloat can occur. Contact allergy is usually manifested by eczema. In farm animals the eczematous lesion is commonly restricted to the skin of the lower limbs, particularly behind the pastern, and at the bulbs of the heels, or to the midline of the back if the allergy is a result of insect bites. In many cases of allergic disease the signs are very transient and often disappear spontaneously within a few hours. Cases vary in severity from mild signs in a single system to a systemic illness resembling anaphylactic shock. On the other hand, cases of anaphylaxis may be accompanied by local allergic lesions.

DIFFERENTIAL DIAGNOSIS

The transitory nature of allergic manifestations is often a good guide, as are the types of lesions and signs encountered. The response to antihistamine drugs is also a useful indicator. Skin test programs as applied to humans should be utilized when recurrent herd problems exist. The differential diagnosis of allergy is discussed under the specific diseases listed earlier.

TREATMENT

Administration of corticosteroids is usually highly effective. Continued exposure to the allergen may result in recurrence or persistence of the signs. Hyposensitization therapy

Amyloidoses

VetBooks.ir

has potential for treatment of recurrent urticaria in horses.12,13 FURTHER READING

Tizard I. Veterinary Immunology. 9th ed. St. Louis: Elsevier Health Sciences; 2013.

REFERENCES

1. Wilson AD. Parasite Immunol. 2014;36:558. 2. Cartwright S, et al. Can J Anim Sci. 2009;89:158. 3. Thompson-Crispi KA, et al. J Dairy Sci. 2012;95:401. 4. Onda K, et al. Vet Rec. 2008;163:418. 5. Holzhauer M, et al. Vet Rec. 2004;154:208. 6. Senturk S, et al. Vet Rec. 2012;170:566. 7. Oryan A, et al. Comp Clin Path. 2010;19:179. 8. Herder V, et al. Vet Microbiol. 2012;155:420. 9. Hurcombe SD, et al. JAVMA. 2007;231:267. 10. Alcott CJ, et al. J Vet Emerg Crit Care. 2010;20:616. 11. Qureshi TA, et al. Int J Pharmacol. 2006;2:357. 12. Rendle DI, et al. Equine Vet Educ. 2010;22:616. 13. Roberts HA, et al. Vet Dermatol. 2014;25:124.

CASEOUS LYMPHADENITIS OF SHEEP AND GOATS SYNOPSIS Etiology Corynebacterium pseudotuberculosis Epidemiology Disease of sheep and goats. Source of infection is discharge from pulmonary or skin abscesses. Infection is through intact skin or skin wounds. Transmission in sheep occurs at shearing and dipping in sheep and in goats and sheep by direct contact. Clinical findings Abscesses in superficial lymph nodes. Respiratory or wasting disease associated with internal abscesses. Clinical pathology Enzyme-linked immunoabsorbent assay (ELISA) tests can be used to determine flock status, but sensitivity and specificity are inadequate to provide reliable identification of infected individuals. Necropsy findings Abscesses in lymph nodes and internal organs. Diagnostic confirmation The clinical and necropsy features are typical. Confirmation is by bacterial culture. Treatment Surgical for superficial abscesses. Control Culling of abscessed sheep or based on serologic testing, hygiene at shearing, avoidance of management risk factors, vaccination.

ETIOLOGY Corynebacterium pseudotuberculosis is the specific cause of the disease. Ovine/caprine isolates are largely a clonal population, distinct from the equine/bovine biotype.1 Both biotypes produce an exotoxin, phospholipidase D, which functions as a sphingomyelinase and is an immunodominant antigen. Variation in toxin production between strains may be related to differences in pathogenicity. The toxic lipid cell wall

mediates resistance to killing by phagocytes and is also a virulence factor. C. pseudotuberculosis is also the cause of ulcerative lymphangitis of cattle and horses and contagious acne of horses, but these have been discussed as separate diseases because they appear to have a separate pathogenesis and do not occur in association with caseous lymphadenitis.

EPIDEMIOLOGY Geographic Occurrence Caseous lymphadenitis occurs in the major sheep-producing countries in the world, including Australia, New Zealand, South Africa, the Middle East, North and South America, the United Kingdom, and most of northern and southern Europe. The disease did not occur in the United Kingdom and the Netherlands until the importation of infected goats in the late 1980s but subsequently spread to be an important disease in both countries. Host Occurrence Caseous lymphadenitis occurs in sheep and goats. Sheep Caseous lymphadenitis increases in prevalence with age and reaches a peak incidence in adults. In one Australian population of unvaccinated sheep the frequency of infection at slaughter was 3.4% for lambs and 54% for adult ewes, and a similar prevalence has been recorded in North and South America. In another large study of mature sheep in Australia the overall prevalence of lesions at slaughter was 26%, with carcass lesions in 20.4% of sheep and offal lesions in 9.5%. The prevalence of infection in ewes culled for age in Western Australia fell from over 50% in the 1980s to approximately 25% in the early 2000s, which was suggested to be partly a result of cessation of compulsory dipping for lice during this period. Following introduction to British flocks in the late 1980s, outbreaks increased to a peak in 1998 but have since decreased. Examination of isolates during this period suggested that all were related to the initial introduction. A serologic survey of 745 flocks showed an overall prevalence of seropositive animals of 10%, with 18% of flocks sampled having one or more positive animals. Goats Prevalence of lesions in goats tends to be lower than for sheep. In domesticated goats an overall prevalence rate of 8% is recorded in the United States, with a similar prevalence recorded in feral goats in Australia. As with sheep, prevalence increases with age and can be as high as 22% at 4 years of age. The assessment of prevalence in goats based on the presence of abscess is complicated by the fact that a significant proportion of abscesses in goats may be

761

produced by Arcanobacterium (Trueperella) pyogenes. Source of Infection The primary habitat of C. pseudotuberculosis is in infected animals. Sources of infection are the discharges from ruptured abscessed superficial lymph nodes and the nasal and oral secretions from animals with pulmonary abscesses draining into the bronchial tree. The organism can survive in pusinfected soil for up to 8 months, in infected shearing sheds for approximately 4 months, and on straw, hay, and other fomites for up to 2 months, but it is not easily isolated from the soil of infected premises. Low temperatures and moist conditions prolong survival time, and infectivity persists in sheep dips for at least 24 hours. Transmission Infection of an animal is facilitated by the presence of skin wounds, but the organism can invade through intact skin. Transmission is by direct contact with infective discharges or contaminated shearing equipment, contaminated shearing shed boards or holding pens, contaminated dipping or shower fluids, or dust from contaminated shearing sheds and yards. Risk Factors Sheep Most studies on risk factors have been conducted in Australia, and observed risk factors may not always apply to management systems in other countries. Age and Sex There is a higher prevalence in older sheep, which probably reflects greater exposure to risk factors such as shearing and dipping. In the United Kingdom a disproportionate number of rams are infected, and there is a significant prevalence of infection in terminal sire breeds, which are an important vector to otherwise closed flocks. The prevalence in rams may be related to the high stocking rate at which rams are kept for most of the year and fighting behavior with transmission through head wounds. Breed All breeds are susceptible, but in New Zealand, which has a mix of fine wool and meat sheep breeds, the prevalence of disease is higher in Merino and Merino-cross breeds. This may relate to greater susceptibility to skin damage at shearing because of their finer skin and the presence of neck wrinkles. In the United Kingdom, infection was initially more prevalent in terminal sire breeds but subsequently spread to hill and upland flocks.2,3 Shearing Shearing is a major risk factor in sheep; in general, infection rates increase with the

762

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

number of times sheep have been shorn. When spread occurs, it occurs mostly within groups of sheep shorn together. Sheep may be infected by transfer of pus from abscesses discharging or cut at shearing, via shearing combs, but spread from sheep with discharging pulmonary abscesses to sheep with skin cuts is considered more important. Close contact of recently shorn sheep in any circumstance may facilitate transmission through contact between infected respiratory secretions and susceptible skin. Sheep are commonly in close contact in collecting pens immediately following shearing, or before dipping, and infected nasal and oral secretions can be deposited directly onto shearing cuts. Keeping sheep under cover for more than an hour after shearing increases the odds for spread. Poor hygiene in the shearing shed, allowing contamination of shearing boards and holding pens, may facilitate infection of sheep. Movement of infection between flocks can occur through contamination of shearing equipment, mobile shearing sheds, or dips and infection on the clothing of shearers. Contract shearing has been shown to be a risk factor in the United Kingdom. Dust Dust from contaminated yards may transmit infection to recently shorn sheep, although epidemiologic studies suggest that environmental contamination is not a major risk factor for disease in Australia. Housing Close contact associated with high stocking rates at pasture or indoor housing for much of the year may lead to high rates of infection. The difference in lesion distribution between sheep in the United Kingdom and Australia is believed to be the result of close contact at shared feed troughs under conditions of intensive husbandry in the United Kingdom. Dips The organism can persist in reused (plunge dip) or recycled (shower dip) fluids used for ectoparasite control. As few as 25 organisms/ mL in the dip can produce infection. Sheep dipped in infected dipping fluid within a few days of having been shorn are especially susceptible to infection because of the ease of contact between the bacteria and the skin, but spread can also occur through dipping sheep shorn 6 months previously. In an experimental study in which infection-free sheep were shorn and exposed to artificially contaminated dips at 0, 2, 4, 8, and 24 weeks after shearing, a larger percentage of the sheep dipped immediately after shearing seroconverted and had lymph node lesions at slaughter. However, lesions also were present at slaughter in sheep dipped 2 or more weeks after shearing, and there was no significant difference in their prevalence in the groups dipped at 2 to 24 weeks after

shearing. This supports the observation that infection can occur through intact skin, possibly in the case of dip-associated infections, influenced by loss of wool grease because of wetting agents in the dip. Shower dipping of sheep immediately after shearing also significantly increases the odds of a high incidence of caseous lymphadenitis. Although shearing and dipping are important risk factors, disease can also be transmitted from sheep with pulmonary abscesses to nonshorn sheep by aerosols. Goats Shearing is not a risk factor, other than with Angoras. The difference in abscess distribution in goats compared with sheep, with a predominance in the head, neck, and sternum in goats, suggests that contact, fomites, and trauma are important vector mechanisms. Social contact, head butting, trauma from browse, and the use of common neck collars and feed troughs are probable risk factors. Pulmonary abscesses are not as prevalent in goats as in sheep and may be of lesser importance as a source of infection. With both sheep and goats, contamination of soil on bedding grounds, in yards, or in shelters may result in persistence of the organism in the environment for periods significant to the transmission of the disease and can result in infection of wounds created by docking and castration and infection in the region of the sternum. Economic Importance In the majority of young infected animals there is no overt clinical disease or impairment of health other than visible abscessation, but the disease is of considerable economic importance to the sheep and goat industries. In sheep, infection has been associated with a 6.6% reduction in clean fleece weight in the first year of infection and a reduction in growth rate. Infection is a significant cause of condemnation of carcass for human consumption, with condemnation rates as high as 3% to 5% for mutton carcasses and 0.02% to 0.03% for lamb carcasses. Condemnation rates and economic loss vary depending on the country, with differences in the number of abscessed lymph nodes dictating condemnation rather than carcass trimming. In goats the hide can represent a significant proportion of the value of the carcass, and blemishes produced by infection markedly reduce hide value. Clinical disease occurs in animals with the disseminated visceral form, which is a cause of reproductive inefficiency, a major cause of the thin ewe syndrome, and a cause of death and culling in older sheep in infected flocks. Zoonotic Implications Human infection is rare, produces a lymphadenitis with a long and recurrent course,

and is an occupational disease of shearers and abattoir workers with infection occurring through cuts. C. pseudotuberculosis may be present in the milk of goats from udders where the mammary lymph node is affected.

PATHOGENESIS

Multiple microscopic abscesses develop in the draining lymph node by 1 day after experimental infection in the skin, and between 3 and 10 days of infection these coalesce to form typical pyogranulomas. The sphingomyelin-specific phospholipidase D exotoxin produced by the organism is believed to facilitate spread of infection by promoting leakage of plasma from small blood vessels at the site of infection, with flooding of lymphatic spaces. Abscesses develop in 60% to 80% of infected sheep. The high lipid content of the bacterial cell wall gives resistance to the digestive enzymes of the phagocyte, and the organism persists as a facultative intracellular parasite. The reduction of wool growth in the first year of infection probably results from the catabolic effects of cytokine and toxic metabolites released during the acute inflammatory and immune response to initial infection. Hematogenous spread of the organism results in abscess formation in many organs, and these may occur in the absence of peripheral lesions. Up to 25% of affected sheep at abattoirs are recorded as having lesions only in thoracic viscera. This tendency for a high incidence of lesions in the lung appears to be general, but prevalence varies considerably between geographic areas. The abdominal visceral and somatic tissues are also commonly affected. Less commonly, hematogenous infection occurs in young lambs to produce septicemic disease.

CLINICAL FINDINGS

There is palpable enlargement of one or more of the superficial lymph nodes. Those most commonly affected are the submaxillary, prescapular, prefemoral, supramammary, and popliteal nodes (Fig. 11-5). The abscesses commonly rupture, and creamy to caseated pus, with no odor, is discharged. Goats have a much greater proportion of lesions in the lymph nodes draining the head, related possibly to superficial injury during browsing. Abscesses may subsequently develop in other lymph nodes. In the United Kingdom, clinical signs of infection in sheep are most commonly associated with the superficial lymph nodes of the head and neck, although in one study over 30% of sheep were found to have visceral lesions.4 Both sheep and goats may also show abscess in the skin, particularly of the face, with loss of overlying hair. In cases in which systemic involvement occurs, chronic pneumonia, pyelonephritis, ataxia, and paraplegia may be present, depending on the site of infection. The debilitating disease of adult ewes commonly

Amyloidoses

763

VetBooks.ir

referred to as thin ewe syndrome is often associated with the occurrence of internal abscesses (81% of ewes), many of which contain C. pseudotuberculosis (86%). Other bacteria, especially Moraxella spp., are also commonly present. In ewes, local spread from the supramammary lymph node to the mammary tissue is common. The resulting fall in milk yields leads to poor growth and even death of lambs, and this may be a serious economic feature in badly affected flocks. Intrascrotal lesions are common in rams but do not involve the testicles or semen.

CLINICAL PATHOLOGY

Fig. 11-5  Cheviot ewes with caseous lymphadenitis of the submaxillary lymph nodes.

A

There is an increase in blood lymphocytes and neutrophils. C. pseudotuberculosis can be cultured from pus obtained by needle biopsy or by transtracheal wash. Serologic tests that have been used in serodiagnosis include indirect hemagglutination, hemolysis inhibition, synergic hemolysis inhibition, immunodiffusion, and ELISA tests to detect antibody to cell-wall antigens or to the phospholipase exoenzyme. Many of these tests have good specificity but few have high specificity and sensitivity. Some have been used to determine flock infection and have been used in eradication schemes.2,3 As yet, none is sufficiently reliable to confidently detect infection in individual sheep. The sensitivity of equivalent tests in goats is generally higher, and they are used for official control schemes in goats in some countries, such as the Netherlands. Radiography and ultrasonography may be useful diagnostic tests in sheep and goats with chronic weight loss and no enlarged external lymph nodes. Radiographs of the thorax may reveal the presence of abscesses in the mediastinal lymph nodes. Ultrasonography of the abdomen may reveal the presence of one or more liver or kidney abscesses. Classic ultrasonographic signs of caseous lymphadenitis abscess is an “onion peel” abscess, which is particularly easy to image in liver parenchyma (Fig. 11-6).

NECROPSY FINDINGS

Caseous abscesses filled with greenish-yellow pus occur chiefly in lymph nodes and to a lesser extent in internal organs. In the early stages the pus is soft and pasty, but in the later stages it is firm and dry and has a characteristic lamellated appearance. Locally extensive bronchopneumonia, with more fluid pus of a similar color, may also be present. Microscopically, nodal architecture is effaced by the abscess. As the lesion expands, the limiting fibrous wall keeps re-forming, creating the “onion-skin” layering noted grossly.

B Fig. 11-6  A, Ultrasonographic image of a liver abscess identified using a 5-MHz sector transducer. The characteristic “onion peel” appearance is highly suggestive of the presence of a Corynebacterium pseudotuberculosis abscess (caseous lymphadenitis). B, Cross-sectional image of the liver from the same sheep obtained at necropsy. Images graciously provided by D. Michael Rings, USA.

Samples for Confirmation of Diagnosis • Bacteriology—lymph node, lung, culture swab from outer portion of abscess (CULT)

764

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

• Histology—formalin-fixed lymph node (LM) DIFFERENTIAL DIAGNOSIS Melioidosis Tularemia Other causes of pneumonia in small ruminants Lymphosarcoma (rare) Suppurative lymphadenitis in lambs has also been found to be associated with infection with Pasteurella multocida, and a disease characterized by the presence of yellow–green pus in abscesses situated in close proximity to the lymph nodes of sheep is associated with a gram-positive micrococcus. The latter disease occurs in France and Kenya and is referred to as Morel’s disease.

TREATMENT The organism is susceptible to antibiotics other than the aminoglycoside group, but treatment is not usually attempted because the abscess is encapsulated, the organism is intracellular, and response is poor. Subcutaneous abscesses can be treated with surgical drainage or extirpation.

CONTROL Culling A measure of control can be achieved by culling all animals with enlarged lymph nodes. Although this is a logical procedure, it is worth noting that it is not capable of detecting early lesions or of detecting those animals with internal abscess but no external abscesses. Ideally, control would be by the identification and culling of infected animals using serologic testing. Culling on the basis of serologic tests has been used in goat herds in the Netherlands, small pedigree flocks in the United Kingdom, and a larger flock of 1000 sheep in Scotland.2,3 The increased sensitivity and specificity of current ELISA tests (87% to 93.5% and 98%, respectively) makes this a feasible strategy, although large numbers of seropositive sheep may be detected and require culling (e.g., 159 following 9 tests in a flock that originally had 108 sheep).2,4 Thus, the cost-effectiveness of test and cull, as opposed to control using vaccination, will be marginal unless the test-and-cull strategy is supported by other funding.2,3 Control of Spread The Mules operation is being gradually phased out in Australian flocks, but all docking implements, ear-taggers, and shears used for this procedure should be dipped in strong disinfectant before each use. Similar attention should be given to the combs and cutters at shearing time. There should be good hygiene and disinfection in the shearing shed, especially of the shearing board and holding pens. Mobile shearing trailers

should be cleaned and disinfected between farms. The importance of personal hygiene should be impressed on shearers, and farmspecific overclothing should be provided if possible. Younger age groups should be shorn first, rams second to last, and any sheep with palpable lesions last. Pus spilled on the shearing floor should be cleaned up and the area disinfected. All shearing cuts should be disinfected. There can still be a high abscess rate in flocks that practice these control procedures. Close contact of sheep following shearing should be avoided. All efforts must also be directed to avoid contaminating dipping fluid; one discharging abscess is capable of contaminating an entire tank of fluid. Dipping after shearing may be undesirable in badly affected flocks. The addition of an efficient bactericidal agent to the dipping fluid is worthy of consideration. Goat housing should be free of wire or other causes of skin trauma, and communal use of equipment such as neck collars should be avoided. External parasites must be controlled. Goat herds that are free of the disease should avoid the purchase of animals from herds with a history of abscessation. Vaccination Vaccines formulated from concentrated, formalin-inactivated C. pseudotuberculosis culture supernatants containing phospholipase D have considerable efficacy and are available in many countries. Attenuated mutant vaccines also show promise. Vaccination does not provide complete protection against the development of abscesses, but controlled field trials show a significant reduction in the number of sheep that develop abscesses and a reduction in the number of abscesses in infected sheep. Vaccinated sheep have fewer lung abscesses than unvaccinated sheep, in one study 96% fewer, and are less likely to spread infection from this source. Compliance with the recommended full course of the vaccine has an important influence on the efficacy of vaccination. An Australian study showed that flocks that followed the recommended protocol of two priming doses to lambs with yearly boosters to adult sheep throughout their life had an average slaughter prevalence of infection in sheep of 3%, whereas the average prevalence of lesioned sheep at slaughter from flocks that only partially followed this protocol, by administering a single dose to lambs or not giving yearly boosters to adult sheep, varied from 22% to 33%. Immunity to caseous lymphadenitis is believed to be associated with antitoxin activity and primarily cell mediated, but colostral immunity will protect against experimental challenge at 6 weeks of life. Colostral immunity will also affect the development of immunity from vaccination, and lambs in flocks with a high prevalence of

caseous lymphadenitis should not be vaccinated at less than 10 weeks of age. Vaccination appears less successful in goats, and although it protects against experimental challenge and spread of the organism from the site of infection, there has been little protection from natural infection in field trials.

PREVENTION

All potential introductions to a flock should be examined clinically for evidence of disease. Although this is not a particularly sensitive method of detection of infection, obvious clinical cases will be detected. Determining the infection status of the source flock is a safer procedure, and if high-level control or eradication is an aim, purchases should be direct and not through markets. The ultimate method of prevention would include serologic testing of individual animal introductions when tests with very a high sensitivity become available.

ERADICATION

Eradication is reported in endemically infected flocks by initial culling of all sheep with clinical signs and subsequent serologic testing and culling of reactors. In one case, seropositive ewes were allowed to lamb before culling but lambs were removed at birth, isolated from the infected dams, and fed cow’s colostrum and milk replacer. These procedures were coupled with rigorous disinfection of facilities, removal of bedding and topsoil from barns and pens, isolation of seronegative sheep for 6 months from previously used pastures and tracks, and hygiene at skindamaging management procedures. Serologically positive sheep were not detected after the second screening.5 Attempted eradication in an extensively managed hill flock in Scotland eliminated clinical disease for 2 years following eight tests during a test-andcull program using an improved ELISA that lasted 2 years, although two seropositive sheep were detected at the last test.3 Total herd or flock eradication and replacement with infection-free animals is also possible, but there is a risk of reintroduction of the disease given that the sensitivity of the current ELISA tests used for screening is only around 90%. The sensitivity and specificity of a bulk milk ELISA were 41.4% and 81.7%, respectively, and thus the screening of bulk milk may be a costeffective way of initially detecting caseous lymphadenitis infection in dairy goats.6 FURTHER READING

Bird GJ, Fontaine MC. Corynebacterium pseudotuberculosis and its role in ovine caseous lymphadenitis. J Comp Pathol. 2007;137:179-210. Radostits O, et al. Caseous lymphadenitis of sheep and goats. In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:795-798. Windsor P. Caseous lymphadenitis in small ruminants. Vet Clin North Am Food A. 2011;27:193-202.

Amyloidoses

REFERENCES

VetBooks.ir

1. Connor KM, et al. Vet Res. 2007;38:613. 2. Baird GJ, Malone FE. Vet Rec. 2010;166:358. 3. Voigt K, et al. Small Ruminant Res. 2012;106:21. 4. Bastos BL, et al. Vet Clin Pathol. 2011;40:496. 5. Schreuder BE, et al. Vet Rec. 1994;135:174. 6 Nagel-Alne GE, et al. Vet Rec. 2015;176:173.

BOVINE FARCY Bovine farcy, a chronic infectious disease affecting the lymph nodes of Zubu cattle; it is endemic in sub-Saharan Africa but has been reported in 19 countries in Africa, Asia, Latin America, and the Caribbean.1–2 In Africa, the disease is caused by Mycobacterium farcinogenes in the eastern and central regions and M. senegalense in the western region. Both species are rapidly growing, gram-positive, branching, and acid-fast mycobacteria. It is not yet certain whether Nocardia farcinica in other parts of the world causes cutaneous nocardiosis (farcy) in animals that mimics bovine farcy.1–2 Epidemiologic data indicate that bovine farcy occurs in adult cattle of the transhumance pastoralist tribes of the Sahel and Sudanian savannah zones. In some areas, 25% to 30% of cattle used to be affected, but the disease has disappeared from many countries where it was once a problem.1 Other domestic and nondomestic animals are not affected. It is not known whether the bacteria are zoonotic, even though an earlier study indicated that the human pathogen Mycobacterium peregrinum type II belongs to the species M. senegalense.3 Ixodid ticks, including A. variegatum, may play a role in disease transmission; breeds of cattle resistant to ticks (e.g., the N’Dama) are resistant to farcy.1 Bovine farcy causes some economic losses as a result of damaged hides and also as a public health burden because the lesions resemble those of bovine tuberculosis in carcasses, and thus the meat from affected animals is considered inappropriate for human consumption. For example, in a study involving 6680 bovine carcasses in Sudan, 400 caseous lesions were identified, only 12 of which were a result of bovine tuberculosis, whereas 59 were caused by bovine farcy.4 The clinical diagnosis of bovine farcy in the late stage of the disease is not difficult because the cordlike nodular lesions in the skin and lymphatics are almost pathognomonic. Laboratory diagnosis is by conventional smears stained with acid-fast stain and by culture. Molecular and serologic tests developed still need to be evaluated for sensitivity and specificity, but the ELISA can be used to support early clinical diagnosis, in epidemiologic surveys, and for screening before animals are exported to farcy-free regions.5 The disease is slowly progressive, and lesions occur in superficial lymph nodes,

mostly in the prescapular and precrural lymph nodes. Affected lymph nodes suppurate, and there is induration of the lymphatic vessels. There can be infection in the mesenteric lymph nodes, with some cases having lesions in the udder or the lung. Lesions are common in areas of the body where A. variegatum ticks attach. Histologic examination shows a severe granulomatous reaction characterized by lymphocyte, macrophage, epithelioid, and giant-cell infiltration, along with marked fibrous proliferation. The agent can be detected in tissue sections by acid-fast staining and by PCR techniques.4 Treatment is not recommended and there is no vaccine. Most cases are detected at slaughter. FURTHER READING Hamid ME. Epidemiology, pathology, immunology and diagnosis of bovine farcy: a review. Prev Vet Med. 2012;105:1. Hamid ME. Current perspectives on Mycobacterium farcinogenes and Mycobacterium senegalense, the causal agents of bovine farcy. Vet Med Int. 2014;247906. doi:10.1155/2014/247906; [Epub 2014 April 30].

REFERENCES

1. Hamid ME. Prev Vet Med. 2012;105:1. 2. Hamid ME. Vet Med Int. 2014;247906. doi:10.1155/2014/247906; [Epub 2014 April 30]. 3. Wallace RJ Jr, et al. J Clin Microbiol. 2005;43:5925. 4. Asil el TA, et al. Trop Anim Health Prod. 2013;45:469. 5. El Hassan HA, Hamid ME. Epud. 2008.

SPORADIC LYMPHANGITIS (BIGLEG, WEED) Sporadic lymphangitis is also called Monday morning disease. It is an uncommon occurrence. It can affect any breed of horse but particularly heavy draught horses. It usually affects single horses, but occasionally outbreaks do occur. There are a few predisposing factors. The most frequent one is rest from work and exercise for 1 to 3 days. Another suspected factor is a change from a low-quality diet to a diet high in protein, such as peas or beans. Both of these may lead to a slowing down of lymphatic flow in the limbs, facilitating ingression of any bacteria trapped on the legs in small wounds or cracks. Dirty stables may also predispose, although the condition is seen in very hygienic stables at times. Continual wet weather may also be a predisposing factor. This is a noncontagious disease of horses characterized by acute fever, lymphangitis, and severe swelling of one or both hindlegs—forelimbs are rarely, if ever, affected. The disease commences abruptly with fever (40.5° to 41° C; 105° to 106° F), shivering, and a rapid pulse rate and respiration. Horses are usually very thirsty. The mouth may be hot and the mucous membranes injected. Lameness rapidly results. Pain in the acute disease can be severe. There is severe pain on palpation of the affected leg, and lameness

765

may be so severe that the horse may refuse to put its foot to the ground. The limb is swollen and hot; the swelling extends from the top of the leg and down to the coronet. There is cording of the lymphatics on the medical aspect of the leg and palpable enlargement of the lymph nodes in some horses. There may be exudate on the skin. Affected legs may pit under pressure. The horses may show perspiration. The acute disease may last only 1 to 3 days, and recovery or conversion into a chronic phase with persistent and variable swelling of the leg, intermittent fever, and variable lameness follows. Occasionally abscesses develop in the lymph nodes and vessels, but usually there is no localization of the infection. There is a tendency for the disease to recur and cause chronic fibrotic thickening of the lower part of the limb extending to the level of the stifle in many horses. Swelling of the leg is often exacerbated by late pregnancy. Horses that have had one attack appear to be prone to further attacks. Recurrences produce a “thick leg,” which is not a result of fluid but of connective tissue. Sporadic lymphangitis can be associated with superficial wounds and ulcers on the lower parts of the limbs, but often there are no wounds detected. The disease is thought to develop as a lymphangitis and, potentially, lymphadenitis of the deep inguinal nodes as a result of these wounds. The affected lymph nodes and swelling of the limb obstruct lymphatic and venous drainage, causing lymphatic obstruction, edema, and, in some cases, cellulitis. Ultrasonographic examination reveals distended lymph vessels that contain fluid that is not echogenic. Ultrasound guided aspiration of this fluid yields fluid with a low total protein concentration and mild neutrophilia (high proportion of the cells present in the fluid are neutrophils, although the absolute count is usually less than 1.0 × 109 cells/L. Culture of the fluid is recommended. Actinobacillus spp., Staphylococcus, Streptococcus, Pasteurella, Pseudomonas, Fusobacterium, and Nocardia have been isolated at times. In many cases there are negative culture results, possibly because the causative agent is difficult to culture, but in some cases Corynebacterium pseudotuberculosis has been isolated, and this appears to be a seasonal occurrence in the late summer and autumn in the United States. The clinical significance of results of culture of the fluid is unknown, but results could be used to guide choice of antibiotics. Radiographic examination is usually unremarkable, apart from demonstrating soft tissue swelling. Affected horses, in both the acute and chronic stages, have mild neutrophilia and hyperfibrinogenemia. It is a difficult disorder to treat effectively. Acutely affected horses should be treated aggressively. The principles of treatment are control of the presumed infection, reduction

766

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

of inflammation, and reduction of swelling. Penicillin or other antimicrobials should be administered parenterally to control the infection. Infection with Corynebacterium requires Rifampin. NSAIDs (flunixin is the first choice, but also phenylbutazone, meglumine, carprofen, or similar) should be administered to control the inflammation and provide pain relief. The limb should be hosed with cold water once to twice daily to reduce heat and provided with gentle massage therapy. Manual massage of the limb might be beneficial. Supportive, compressive bandaging of the limb can reduce the swelling. The horse should be exercised as much as is practical and humane. Horses with chronic disease should be treated with prolonged courses of antimicrobials (penicillin, sulfonamide–trimethoprim combinations, enrofloxacin, or rifampin in combination with sulfonamide–trimethoprim), nonsteroidal drugs, and local therapy. Acute exacerbations can be managed by administration of dexamethasone (40 µg/ kg orally or parenterally, once daily for 5 days, and then gradually tapering). This dose is not abortifacient in pregnant mares. Exercise and supportive bandaging are important in minimizing the swelling. The chronic disease requires prolonged and intermittent therapy, often for the rest of the horse’s life. Differential diagnoses include cellulitis (inflammation of the connective tissues) and ulcerative lymphangitis. Ulcerative lymphangitis is often accompanied by cording of the lymphatics, the formation of hard nodules, and the occurrence of abscesses with a discharge of greenish fetid fluid. Epizootic lymphangitis caused by Histoplasmosa farciminosum in Asia and the Mediterranean is a differential diagnosis. In the Middle East, Eastern Asia, Eastern Europe, and North Africa, Glanders is also a possibility. Sporotrichosis (Sporothrix schenckii) lymphangitis can also be a differential diagnosis. Prevention of the disease necessitates prompt and careful treatment of all wounds of the lower limbs. Provision of daily exercise, restriction of the diet during prolonged rest periods, and dry standing in the stable also help to prevent the disease. Animals compelled to stabling should have a reduced diet, with corn being replaced by bran mash. The legs should be kept clean and disinfected if animals are stabled. Animals affected with the chronic condition should be kept constantly at reasonable work.

TICK-BORNE FEVER (ANAPLASMA PHAGOCYTOPHILA) SYNOPSIS Etiology Anaplasma phagocytophilum Epidemiology Occurs in the northern latitudes and is transmitted by Ixodes

ricinus in United Kingdom and Europe and Ixodes scapularis and Ixodes pacificus in the United States. Disease in sheep and cattle primarily reported from the United Kingdom and Europe. Seasonal occurrence associated with the feeding activity of the vector. More severe disease in naive introduced animals. Increases susceptibility to other infections, especially in sheep. Clinical findings Fever, depression, lethargy, polypnea, and fall in milk production in cattle. Abortion. Clinical pathology Thrombocytopenia followed by more prolonged neutropenia and lymphocytopenia. The organism is demonstrable in the neutrophils and monocytes during each febrile period. Diagnostic confirmation Demonstration of the A. phagocytophilum in leukocytes at acute stage of the disease or by serology retrospectively. Treatment Oxytetracycline. Control Oxytetracycline during risk period. Tick control.

ETIOLOGY Tick-borne fever (also called pasture fever in cattle) is a tick-transmitted disease of sheep and cattle in the northern hemisphere and is caused by Anaplasma phagocytophilum (formerly Ehrlichia phagocytophilia, Cytoecetes phagocytophila, and Ehrlichia equi). The genus Anaplasma (Rickettsiales: Anaplasmataceae) contains obligate intracellular gramnegative bacteria found exclusively within membrane-bound inclusions or vacuoles in the cytoplasm of both vertebrate and invertebrate (tick) host cells. The genus includes A. marginale, A. centrale, A. bovis, and A. ovis which are pathogens of ruminants; A. phagocytophilum, which affects a wide range of hosts, including humans, wildlife, and domestic animals; and A. platys, which infects dogs.1 A. phagocytophilum replicates primarily in the cytoplasm of neutrophils and is 0.4 to 1.3 um in size. The life cycle is biphasic, transitioning between the noninfectious reticulate cell that replicates and the smaller dense core form that is infective to mammalian and tick cells.2–3 There are strains (variants) of A. phagocytophila that have biological and ecological differences, including variations in host pathogenicity, vectors, and geographic distribution. Although the mechanisms for these differences remain largely elusive,4–5 the type IV secretion system (T4SS) may play a role in strain virulence of A. phagocytophilum.6 In sheep, different variants of A. phagocytophilum may exist simultaneously in the same sheep flock. Tick-borne fever is also used as a name for similar, but less well-defined, diseases of ruminants that are associated with infection with related organisms such as Anaplasma (Ehrlichia) bovis. These are reported from

other areas of the world, such as India and Africa, and are transmitted by the ticks Rhipicephalus appendiculatus, Amblyomma variegatum, and Hyalomma truncatum. Currently, A. phagocytophilum is viewed as a single bacterial species capable of infecting a broad range of hosts and is based on 16S rRNA gene analysis.7 The description that follows is of tick-borne fever of sheep and goats and pasture fever of cattle associated with A. phagocytophilum.

EPIDEMIOLOGY Occurrence Infection with A. phagocytophilum occurs in a wide range of mammalian hosts, including humans, dogs, sheep, cows, horses, wild deer, and rodents. The association of A. phagocytophilum with human granulocytic ehrlichiosis in the mid-1990s has led to much activity in defining its geographic occurrence by serologic surveys or detection in ticks by molecular methods. These studies have determined that the organism is present where the host ticks are present in Europe, North America, the Middle East, and Asia.4 A. phagocytophilum is arguably the most widespread tick-borne infection in animals in Europe.8 However, the disease tick-borne fever, as opposed to infection, occurs primarily in certain areas of the United Kingdom, Ireland, Norway, Finland, France, Germany, Spain, and Switzerland. Because ticks favor particular optimal environmental conditions, the geographic distribution of the ticks is usually restricted to a specific area (small or large), and tick-borne fever only occurs in these areas. Within these areas infection can be intense; in the endemic coastal area of Norway, close to 100% of sheep grazing Ixodes-infested pastures are infected. Tick-born fever has a seasonal occurrence in association with the feeding activity of the vector tick. Infection can be endemic in affected areas. Sheep, cattle, goats, deer, and reindeer may be infected. The disease has long been known as a disease of sheep but in recent years is being recognized as a common infection in cattle in at-risk areas. The incidence rate of infection is high, but clinical disease may be mild and not easily observed in many areas where this disease occurs because these areas are commonly wild with little human habitation and little frequent observation of at-risk livestock. Infection, as determined by seropositivity, can occur in sheep that have had no clinical evidence of disease because of the existence of variants with low pathogenicity. The disease in horses, previously known as Equine Granulocytic Ehrlichiosis, is described separately in this chapter as “Equine anaplasmosis”. Source of Infection and Transmission In Europe, A. phagocytophilum is transmitted by the three host tick Ixodes ricinus,

Amyloidoses

VetBooks.ir

which requires a single blood meal at each stage of development. The tick feeds for approximately 3 weeks every year and completes its life cycle in 3 years. The larval and nymphal stages will feed on any vertebrate, but the adult female will engorge and mate only on larger mammals. A. phagoctyophilum infects and multiplies in the epithelium of the midgut and salivary glands of ticks, from which it is transmitted to vertebrate hosts during feeding. A tick becomes infected by feeding on an infected host, and there is transstadial but not transovarial passage of the organism. It is estimated that the majority of ticks are infected with the organism in enzootic areas, and one former study of ticks from a field site found 44% of nymphs and 32% of adults infected but no infected larval stages. There is a close relation between tick density and the proportion of sheep and ticks infected with A. phagocytophilum, but it is nonlinear and complex. In the United States, Ixodes scapularis has been implicated in transmission of the organism in eastern United States and Ixodes pacificus on the West Coast, as have Ixodes persulcatus and Haemaphysalis longicornis in Asia, but clinical disease in ruminants is not a feature in these locations. Few cases of congenital infection of calves have been reported,9 and the organism is also present in leukocytes in milk during the acute phase of the disease, but the significance of this in the epidemiology of the disease is not known. As few as one A. phagocytophila–infected cell may be enough to transmit infection, and use of a single needle between sheep in a group could possibly transmit infection. It has been suggested that the presence of ticks in migratory birds could spread infection of this agent to other geographic regions.10–11 Experimental Reproduction The disease can be readily reproduced experimentally. The severity of the clinical response of sheep following experimental infection is not dose dependent, and there is no dose effect on the degree of bacteremia or neutropenia. Host Risk Factors Calves and lambs are much more susceptible than adults, although clinical disease may be less severe in very young lambs, possibly because of the mitigating effects of colostral antibody. Hyperimmunization of the pregnant ewe will produce high levels of colostral antibody that will protect the lamb against experimental challenge. However, in the field, colostral immunity does not protect against infection, and lambs born of ewes raised in endemic areas become infected. Natural infection is followed by a state of low-grade premunity as a result of the presence of the organism in the blood, which

provides partial resistance to subsequent infections, with the disease manifesting itself in a less severe form. Once infected, animals probably remain carriers for life and act as reservoirs of infection for new generations of ticks. The case-fatality rate is very low, and most reported mortality is in association with intercurrent disease. A significant indirect effect of tick-borne fever is that it increases the susceptibility of lambs to staphylococcal pyemia, staphylococcal pneumonia, septicemic and pneumonic pasteurellosis, louping-ill, and possibly other diseases. The mortality rate is negligible in cattle but may be higher in sheep. Pathogen Risk Factors The activity of the tick is seasonal, and consequently tick-borne fever has a seasonal occurrence. The tick is active at temperatures between 7° (44° F) and 18° C (64° F), and most ticks feed in the spring, with peak activity dependent on the latitude and elevation of the pasture but generally occurring in April and May. In some areas there is a second period of activity of a separate population of I. ricinus in the autumn during August and September. Clinical signs in cattle occur predominantly in spring, 1 to 2 weeks after they start to graze. Zoonotic Implications Human granulocytic anaplasmosis (HGA) is associated with A. phagocytophilum and was first described in the United States in 1994 and is now regarded as an emerging disease. There are few reports from Europe, Russia, and Japan.4 Clinical and laboratory findings are fever, myalgia, headache, malaise, thrombocytopenia, leukopenia, anemia, mild hepatic injury, and splenomegaly, with symptoms varying from none to mortality.12 The disease presents most commonly as an undifferentiated, febrile, potentially severe illness occurring in summer or spring associated with occupational or recreational activities that allow exposure to infected ticks. There is no recognized direct zoonotic risk from exposure to infected animals, but the mammalian reservoir for A. phagocytophilum infection within the United States includes several rodents, foxes, and possibly other wild animals. These are infected with the human pathogenic variant (Ap-ha). Other animals, including the white-tail deer, are infected with different strains of the bacterium not infective to humans (Ap-V1).4,7 The organism is also present in leukocytes in milk during the acute phase of the disease, but the risk to humans consuming infected milk is probably minimal because strains of A. phagocytophilus differ in host infectivity.4

PATHOGENESIS

A. phagocytophilum infects and replicates within the neutrophil, but can also infect

767

endothelial cells. The organism has evolved the remarkable ability to subvert or hijack the powerful innate antimicrobial defenses of host cells.4 For example, A. phagocytophilum creates an intracellular membranebound compartment that allows replication and nutrition in seclusion from lysosomes and thereby suppresses respiratory burst.13 Furthermore, it subverts the innate immune system of neutrophils by inhibiting apoptosis (through activation of an antiapoptosis cascade) and by inducing autophagy, both to keep the host cell alive and to create a safe haven for replication.4,14–17 In addition, individual animals that survive acute infection develop persistent infection lasting for several months or even for life. The persistence of infection and recurrent bacteremia is a result of antigenic variation thought to be mediated by the bacterial major surface protein 2 (MSP2/p44).18 Furthermore, endothelial cells of the microvasculature may play a crucial role in the development of persistence because they may be an excellent site for dissemination of the organism to circulating neutrophils.19 Tissue pathology is not associated with direct A. phagocytophilum–mediated injury but results from immunopathologic mechanisms associated with activated macrophages and elevation of levels of the proinflammatory cytokine, interferon gamma (IFN-y), and chemokines.12,20,21 Fever develops in association with bacteremia and is the prominent clinical abnormality in the experimental disease. It persists for approximately 8 days, may exceed 41° C (105.8° F), and is accompanied by depression. Although this syndrome is of limited importance in the experimental setting, the occurrence of fever, dullness, and depression of the sucking drive can be a significant influence on the viability of lambs in the cold, wet, rough-grazing areas where this disease commonly occurs and may contribute to lamb mortality. Tick-borne fever produces profound effects on immunologic defense systems. By impairing the function of neutrophils, the bacterium causes infected animals to become more susceptible to opportunistic infections. There is also a prolonged neutropenia lasting for 2 to 3 weeks combined with a thrombocytopenia. Up to 70% of the neutrophils are infected from the onset of the bacteremia. There is significant lymphocytopenia that develops 6 days after infection and that affects all T- and B-lymphocyte subsets. The antibody response of infected sheep to immunogens such as tetanus toxoid is also impaired. Field observations and experimental challenge have shown that infected lambs are more susceptible to disease and mortality from intercurrent infections. The ability of an infection with A. phagocytophilum to predispose to secondary disease varies with the strain of the organism, which may explain

768

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

why secondary complications are not observed in all flock infections with tickborne fever. There is a clear relationship between infection with A. phagocytophilum and susceptibility to infection with S. aureus and the resultant disease tick pyemia. This is established both by epidemiologic and experimental studies. Concurrent infection of sheep with the agent of tick-borne fever potentiates the pathogenicity of louping-ill virus in experimental infections, resulting in more severe disease and a higher mortality rate. Both diseases are transmitted by I. ricinus. However, in areas where both diseases are endemic, colostral immunity will delay infection of lambs with the louping-ill virus until the second year of exposure to the vector tick while allowing infection with tick-borne fever. Simultaneous primary infection with both agents may be uncommon in nature. Infection also facilitates invasion and systemic mycotic infection with Rhizomucor pusillus, resulting in diarrhea and dysentery and a high mortality rate. Concurrent experimental infection of sheep with A. phagocytophilum and Chlamydia psittaci results in chlamydial pneumonia, and simultaneous infection with parainfluenza-3 (PI-3) virus potentiates the pathogenic effect of PI-3 virus. The immunosuppressive effect of tickborne fever is believed to have resulted in the exacerbation of latent Brucella abortus in a naturally occurring abortion outbreak in cattle. Concurrent infection of tick-borne fever and Listeria monocytogenes or Pasteurella hemolytica promotes the respective septicemic disease in lambs.

CLINICAL FINDINGS

Disease is generally benign, but infection can produce abortion and can cause significant loss of weight in lambs and calves. Cattle In cattle, there is an incubation period of 5 to 9 days followed by a rise in temperature to about 40.5° C (105° F), which persists for 2 to 12 days and for a longer period in late pregnant cows than in lactating cows. The temperature falls gradually and is followed by a secondary febrile period and, in some cases, yet further episodes of pyrexia. During each febrile period there is a marked fall in milk yield, lethargy, polypnea, and, in experimentally produced cases, a mild cough, although feed intake is not reduced. The fall in milk production can be severe and may be the first indication of infection.30 Pregnant cattle in the last 2 months of pregnancy and placed on tick-infected pastures for the first time commonly abort, and occasionally animals die suddenly. The abortions occur shortly after the systemic disease. Some calves are born alive, but they are weak and soon die. In a recent outbreak affecting four dairy cows in Germany, clinical findings included high fever, decreased milk production, lower

limb edema with stiff walking, eye and nasal discharge, and depression. These signs developed about a week after the animals had been brought to the pasture for the first time. All cows recovered after 5 to 15 days, although DNA of A. phagocytophilum could be detected by real-time PCR (qPCR) up to 6 weeks after onset of the disease.22 Sheep In sheep, the syndrome is similar to that observed in cattle, except that respiratory distress is not observed. However, there can be marked differences in clinical manifestation, neutropenia, and antibody response with different variants of A. phagocytophilum. An experimental infection of mature sheep in western United States generally resulted in a subclinical disease.23 Conversely, sheep experimentally infected in the United Kingdom developed primary bacteremia (bacterial DNA) lasting for over 2 weeks and accompanied by fever, followed by secondary and recurrent cycles of bacteremia each lasting for 1 to 3 days without fever.24 The sheep remained persistently infected for up to 358 days. The red deer may be a reservoir for sheep in Norway.25 The reaction in young lambs is quite mild and manifested only by a fever, which fluctuates between 40° (104.0° F) and 42° C (107.6° F) for up to 10 days. Ewes exposed to the disease for the first time commonly experience outbreaks of abortion, and affected rams are temporarily infertile. Abortion is a major manifestation in northern Spain, whereas in the Scandinavian countries the main consequence of infection is immunosuppression leading to secondary infections with S. aureus (tick pyemia) and P. hemolytica. Goats Tick-borne fever in goats is characterized by high fever, dullness, and tachycardia. Horses See the section on equine granulocytic anaplasmosis later in this chapter for a description of the disease in horses.

CLINICAL PATHOLOGY

At the commencement of the fever there is severe but transient thrombocytopenia, and this is followed by more prolonged neutropenia and lymphocytopenia. The anaplasmae are demonstrable in the neutrophils and monocytes during each febrile period and for a few days afterward in cattle and for several weeks in sheep; they can be detected as intracytoplasmic inclusion bodies or morulas in Giemsa-stained blood smears and confirmed by electron microscopy or PCR.26 Several PCR techniques (conventional, nested, or real-time) for the identification of A. phagocytophilum infection in blood and tissue samples have been established, primarily on the basis of the 16S rRNA, groEL,

and p44 genes.7 In China, the rapid and simple loop-mediated isothermal amplification (LAMP) assay targeting the msp2 gene of A. phagocytophilum was found to have a high level of sensitivity comparable to that of nested PCR and qPCR for the detection of the organism in rural human patients.27 Serologic diagnosis is possible using counterimmunoelectrophoresis, which detects IgM antibody, or indirect immunofluorescence using cytospin preparations of blood granulocytes, which detects IgG. Antibody is at a high level at the second week after experimental infection with both tests and is detectable for 6 to 8 weeks with counterimmunoelectrophoresis and for at least 18 weeks with the indirect fluorescent antibody test. ELISA is also available for serologic diagnosis. A commercial ELISA kit is available for rapid in-house identification of A. phagocytophilum antibodies in dogs, but the kit can be used for horses and possibly sheep.28–29 Transmission of the disease for diagnosis may be affected by the IV injection of blood taken at the height of the fever.

NECROPSY FINDINGS

An enlarged spleen, up to 4 to 5 times the normal size, with subcapsular hemorrhages is indicative of tick-borne fever in sheep in endemic areas. Histologically, the only characteristic lesion is a depletion of lymphocytes from lymphoid tissues. Multifocal leukomalacia (spongy change of white matter) and swelling of oligodendrocytes have been reported in the brain of aborted lambs, probably the result of fetal anoxia. DIFFERENTIAL DIAGNOSIS The geographic restriction of the disease and its relation to tick infestation are diagnostic features, but the clinical signs are quite nonspecific. Lesions attributable to concurrent bacterial, mycotic, and viral infections may overshadow the primary disease in sheep and are more likely to be the cause of death. The disease in cattle has some similarity to bovine petechial fever (Ondiri disease), associated with E. ondiri, which occurs only in Kenya.

TREATMENT TREATMENT AND CONTROL Treatment Long-acting tetracycline (20 mg/kg IM at early stages) (R1) Oxytetracycline (10 mg/kg IV daily for 5 days at early stages) (R1) Control Long-acting tetracycline (20 mg/kg IM to lambs and calves at risk) (R2)

Amyloidoses

VetBooks.ir

The best results are with tetracyclines, although cattle may recover without therapy. In sheep, a single dose of long-acting tetracyclines (20 mg/kg IM) or a 5-day course with oxytetracycline (10 mg/kg IV daily) given during the acute phase of the disease is effective in treatment, but infection is not eliminated in a significant proportion of sheep. In goats, good results are provided by a single dose of oxytetracycline (10 mg/kg BW IV). A potentiated sulfonamide containing trimethoprim and sulfadimidine, and sulfamethylphenazole (20, 50, and 50 mg/kg, BW respectively) were used in the past; ampicillin is ineffective. The Anaplasma organisms persist in treated animals, which may subsequently suffer a relapse.

CONTROL

Control of tick-borne fever depends on control of the tick population. The annual dipping of ewes with organophosphates or synthetic pyrethroid acaricides will help reduce tick numbers, and the double dipping of lambs during the tick season will help reduce disease in the lambs but can be difficult to achieve in the terrain and with the management practices of affected areas. Disease is reduced if the flock can be kept off the tick-infested pastures until the lambs are 6 weeks old and if the flock is dipped before introduction to the pasture. In some areas it may be possible to reduce tick numbers by pasture management systems that disturb the pasture microclimate required by the tick. In Norway, frequent pour-on applications of pyrethroids in lambs reduced tick infestation rate but did not reduce the seroprevalence of A. phagocytophilum on tickinfested pasture.30 Furthermore, frequent use of chemical acaricides has led to growing concerns about environmental safety, human health, increasing costs, and increasing resistance of ticks to pesticides. Studies on the biological control of ticks are ongoing. Vaccines against ticks and/or against A. phagocytophilum are also being studied. The disease can be more severe when adult animals are exposed to infection for the first time, and naive late-pregnant cattle should not be introduced to tick-infested pastures during the tick-rise periods. A single administration of 20 mg/kg of longacting tetracycline is reported to provide protection against experimental challenge for periods up to 3 weeks. The prophylactic administration of long-acting tetracycline to lambs and to calves during the season of tick activity is reported to reduce mortality and improve growth rates over untreated controls. FURTHER READING De la Fuente J, et al. Functional genomics and evolution of tick-Anaplasma interactions and vaccine development. Vet Parasitol. 2010;167:175. Rikihisa Y. New findings on members of the family Anaplasmataceae of veterinary importance. Ann NY Acad Sci. 2006;1078:438.

Rikihisa Y. Mechanisms of obligatory intracellular infection with Anaplasma phagocytophilum. Clin Microbiol. 2011;24:469. Rikihisa Y, Lin M, Niu H. Type IV secretion in the obligatory intracellular bacteria Anaplasma phagocytophilum. Cell Microbiol. 2010;12:1213. Severo MS, et al. Anaplasma phogocytophilum: deceptively simple or simply deceptive? Future Microbiol. 2012;7:719. Stuen S. Anaplasma phagocytophilum—the most widespread tick-borne infection in animals in Europe. Vet Res Commun. 2007;(suppl 1):79. Stuen S, et al. Anaplasma phagocytophilum—a wide multihost pathogen with highly adaptive strategies. Front Cell Infect Microbiol. 2013;3:31. Woldehiwet Z. Anaplasma phagocytophilum in ruminants in Europe. Ann NY Acad Sci. 2006;1078:446. Wolderhiwet Z. Immune evasion and immunosuppression by Anaplasam phagocytophilum, the causative agent of tick-borne fever of ruminants and human granulocytic anaplasmosis. Vet J. 2008;175:37.

REFERENCES

1. Estrada-Pena A, et al. BMC Biol. 2009;7:57. 2. Troese MJ, et al. Infect Immun. 2011;79:4696. 3. Kahlon A, et al. Infect Immun. 2013;81:65. 4. Severo MS, et al. Future Microbiol. 2012;7:719. 5. Rikihisa Y. Clin Microbiol Rev. 2011;24:469. 6. Al-Khedery B, et al. BMC Genomics. 2012;13:678. 7. Stuen S, et al. Front Cell Infect Microbiol. 2013;3:31. 8. Stuen S. Vet Res Commun. 2007;(suppl 1):79. 9. Henniger T, et al. Acta Vet Scand. 2013;55:38. 10. Geller J, et al. Vector Borne Zoonotic Dis. 2013;13:443. 11. Kang JG, et al. Vector Borne Zoonotic Dis. 2013;15. 12. Chen G, et al. Infect Immun. 2012;89:3194. 13. Woldehiwet Z. Vet J. 2008;175:37. 14. Ayllon N, et al. Infect Immun. 2013;81:2415. 15. Niu H, Rikihisa Y. Autophhagy. 2013;9:787. 16. Niu H, et al. Proc Natl Acad Sci USA. 2012;109:20800. 17. Sarkar A, et al. Infect Immun. 2012;80:1615. 18. Thomas RJ, et al. Vet Microbiol. 2013;pii: S0378. 19. Wang J, et al. Med Microbiol Immunol. 2015;[Epub ahead of print]. 20. Woldehiwet Z, Yavari C. J Comp Pathol. 2014;150:351. 21. Choi KS, Dumler JS. Microbiol Immunol. 2013;57:207. 22. Nielder M, et al. Tierarzti Prax Ausg G Grosstiere Nutztiere. 2012;40:101. 23. Gorman JK, et al. Am J Vet Res. 2012;73:1029. 24. Thomas RJ, et al. J Comp Pathol. 2012;147:360. 25. Stuen S, et al. Ticks Tick Borne Dis. 2013;4:197. 26. OIE manual of diagnostic tests and vaccines for terrestrial animals, World Animal Health Organisation 6th ed; 2008; Chapter 2.4.1. 27. Pan L, et al. J Clin Microbiol. 2011;49:4117. 28. Granquist EG, et al. Vet Immunol Immunopathol. 2010;133:17. 29. Hansen MG, et al. Acta Vet Scand. 2010;52:3. 30. Stuen S, et al. Acta Vet Scand. 2012;54:31.

ANAPLASMOSIS DUE TO A. MARGINALE AND A. OVIS SYNOPSIS Etiology Anaplasma marginale, a rickettsial bacterium in cattle and wild ruminants, and

769

A. ovis in sheep and goats. A. centrale causes mild anaplasmosis in cattle. Epidemiology Common in tropical and subtropical regions; sporadic in temperate regions. Carrier animals are the source of infection. Disease transmitted by ticks, mechanically by tabanid vectors, iatrogenically, and transplacentally. Disease can be endemic in tick areas or sporadic in interface regions between endemic and free areas. Clinical findings In cattle, death or severe debility, emaciation, anemia, and jaundice are the major clinical signs. The disease is usually subclinical in sheep and goats. Clinical pathology Anemia, demonstration of organism in red cells by microscopy, fluorescent stains or polymerase chain reaction (PCR), serology. Necropsy findings Anemia and attendant findings. Demonstration of organism. Diagnostic confirmation Detection of the organism in blood smears, positive serology, PCR, and in some circumstances positive transmission tests. The sensitivity in a group of animals can be increased by using parallel blood smears, serologic tests, and PCR tests. Treatment Clinical cases treated with tetracycline, imidocarb, or enrofloxacin. Blood transfusion. Carrier state not readily eliminated by treatment with tetracycline. Control Tetracycline provides temporary or prolonged protection in face of an outbreak. Vaccination with killed A. marginale vaccine or live A. centrale vaccine used in endemic areas along with vector control. In nonendemic areas, serologic identification of carriers and culling or treatment of reactors. Prevention of iatrogenic transmission.

ETIOLOGY The genus Anaplasma (Rickettsiales: Anaplasmataceae) contains obligate intracellular gram-negative bacteria found exclusively within membrane-bound inclusions or vacuoles in the cytoplasm of both vertebrate and invertebrate (tick) host cells. The genus includes A. marginale, A. centrale, A. bovis, and A. ovis, which are pathogens of ruminants; A. phagocytophilum, which affects a wide range of hosts, including humans, wildlife, and domesticated animals; and A. platys, which infects dogs.1 A. marginale is the type species of the genus; it is transmitted by ticks and other vectors and was first described by Sir Arnold Theiler in South Africa at the beginning of the twentieth century. A. marginale is the causative agent of anaplasmosis in cattle, buffalo, and wild ruminants, and A. ovis in sheep and goats. A. centrale is closely related to, or a subspecies of, A. marginale and causes mild anaplasmosis in cattle. It was originally isolated in Africa but has been introduced as an

770

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

immunizing agent in Australia, South America, and Asia. Molecular studies have identified and characterized several major surface proteins (MSPs) in A. marginale involved in interactions of the organism with both vertebrate and invertebrate hosts.1–2 For example, MSP1a is involved with adhesion to bovine erythrocytes and to tick cells, and it can be used as a genetic marker for identifying strains of A. marginale.3–4 Many geographic strains have been identified worldwide, and they vary in genotype, antigenic composition, morphology, and infectivity for ticks.1–2 Another major surface protein, MSP2 is thought to be involved in antigenic variation to evade the mammalian host immune response, and it is also involved with infection and survival in the tick vector, thus contributing to maintenance of persistent infections in both hosts.1,5 Yet another protein, MSP4 is a stable marker for the genetic characterization of strains and does not undergo antigenic variation when cycling between tick and mammalian hosts.6 As for A. ovis affecting sheep, goats, and some wild ruminants, the MSP4 is also used for genetic characterization and differentiation from A. marginale and other rickettsial organisms.7 Mongolian reindeer are also affected.8

EPIDEMIOLOGY Geographic Occurrence Anaplasmosis in cattle is common and worldwide in distribution, being present on all six continents, but at varying degrees even within countries. It is transmitted through tick bites or by the mechanical transfer of fresh blood from infected to susceptible cattle from biting flies or by bloodcontaminated fomites, including needles, ear-tagging, dehorning, and castration equipment.9 Infection in cattle is endemic in tropical and subtropical areas that support large populations of these vectors, and prevalence rates as high as nearly 80% have been reported in cattle10–12 and 40% in buffalo.13 Infection occurs more sporadically in temperate-climate areas where vectors are seasonal; prevalence rates can be as low as 15% in Iowa14 and 0% to 2% in Canada.15 In the United States and in other countries, the disease has occurred beyond the boundaries of tick-infested areas. Whereas anaplasmosis is enzootic throughout the southern Atlantic states, the Gulf Coast states, and many of the midwestern and western states, the disease occurs sporadically in the northern states and extends to the Canadian provinces of Saskatchewan, Manitoba, Ontario, and Quebec.15 In Europe, anaplasmosis is endemic in the Mediterranean countries of Italy, Spain, and Portugal and has been advancing northward in recent years, with sporadic cases in France, Switzerland, the Netherlands, Hungary, and Austria. A clinical case

of A. centrale infection has been reported in Italy.16 In Australia infection is closely related to the distribution of Boophilus microplus, which is restricted to the northern areas. Prevalence rates are negligible in cattle south of the tick line, but above the tick line the rates increase from south to north. Differences in enzootic and epizootic areas in South America and Africa are also largely related to tick distribution and climate. In most countries there is wide geographic variation in prevalence rates, and this variability contributes to the development of geographically stable or unstable enzootic regions. There is concern, and some evidence, that the global warming trend will expand the boundaries and movement of host ticks. Anaplasmosis of sheep and goats has a distribution similar to that of cattle, and in endemic areas the prevalence rate can be up to 100%.17–18 Host Occurrence Cattle and buffalo are susceptible to A. marginale and A. centrale and sheep to A. ovis. A. marginale will establish in sheep by experimental infection, but A. ovis will not infect cattle. A variety of species of wild ruminants can be infected and may have significance as reservoirs for A. marginale. In the United States the black-tail deer (Odocoileus hemionus columbianus) in the West Coast region is believed to be a reservoir. In Canada, six of six free-ranging mule deer (Odocoileus hemionus) from British Columbia tested positive for A. marginale by PCR,19 and in Brazil, 79.3% of free-living and captive brown brocket deer (Mazama gouazoubira) and marsh deer (Blastocerus dichotomus) tested positive.20 A number of species of antelope in Africa and deer in Europe play a similar role. As for A. ovis, potential wildlife reservoirs include the bighorn sheep (Ovis canadensis) and the mule deer in western United States,7 the farmed white-tail deer in Indiana,20 and the roe and red deer in Europe.21–22 Source and Methods of Transmission The source of infection is always the blood of an infected animal. Recovery from acute infection results in persistent infection characterized by repetitive cycles of rickettsemia. Persistent carriers are the reservoir for herd infection. The level of parasitemia is often too low for detection by microscopy but can be detected by nucleic acid probe analysis. Transmission is biologically by ticks but can also occur transplacentally. Mechanical transmission is by biting flies or bloodcontaminated fomites. Hematophagous Insect Transmission Spread from animal to animal occurs chiefly by insect vectors. A variety of arthropods may act as vectors, but significant natural

vectors are ticks in the family Ixodidae and flies in the family Tabanidae. Of the ticks, the one-host Rhipicephalus (Boophilus) spp. are of major importance in tropical and subtropical regions, and the three-host Dermacentor spp. are of major importance in the western United States. The organism undergoes a complex developmental cycle in the gut cells of ticks, and the final infective stage is present in the salivary gland. Transstadial transmission of the organism occurs in ticks, but there is little evidence for transovarial transmission of A. marginale; however, it has been reported for A. platys.23 Intrastadial transmission is significant with some species, and transmission occurs as the ticks move from one host to another while they are engorging, including from cow to calf. Male D. andersoni can act as effective vectors in this manner for at least 120 days. There appears to be no developmental sequence of Anaplasma spp. in flying insects. Tabanids are efficient mechanical vectors and can transmit infection for 2 hours after feeding. Sucking lice (Haematopinus spp. and Linognathus spp.) have been identified as potential vectors of anaplasmosis in cattle, goats, and buffalo.24–25 The sheep keg (Melophagus ovinus) and deer keg (Lipoptena cervi) are also potential mechanical vectors of A. ovis in sheep and deer, respectively.22 Nevertheless, tick-borne biological transmission is probably most important and is at least two orders of magnitude more efficient than mechanical transmission by flies26 because the bacteria are able to undergo cyclical development and multiplication only in the tick. Over 20 species of tick have been incriminated as vectors worldwide. In Australia the ticks Boophilus microplus and Rhipicephalus sanguineus are the vectors, and in South Africa it is B. microplus, B. decoloratus, and Rhipicephalus simus. In the United States Boophilus annulatus, Dermacentor andersoni, D. variabilis, Argas persicus, biting flies of tabanid species, and eye gnats (Hippelates pusio) also act as vectors. The male ticks of Dermacentor albipictus (the winter tick) and D. occidentalis (the Pacific Coast tick) parasitize both deer and cattle and have been suspected as vectors. D. reticulatus is widely distributed in Europe, from the British Isles to Central Asia, and the males have been shown to be competent vectors.6 Iatrogenic Transmission Anaplasmosis may also be spread mechanically by infected hypodermic needles; by castrating, spaying, tattooing, ear-tagging, and dehorning instruments; and by blood transfusions and embryo transplants. In one study, iatrogenic transmission was detected in 6 of 10 steers sham-vaccinated with a needle fitted to a multiple-dose syringe.27 The ease with which the infection is spread mechanically may vary with the virulence of

Amyloidoses

VetBooks.ir

the rickettsial strain, and this method of spread may be more important in some countries than others. Anaplasmosis may also be spread when cattle, used as donors of infected blood for immunization against babesiosis, are carriers of A. marginale, with the reaction occurring some 3 weeks later than that resulting from the babesia. Transplacental Transmission Intrauterine infection also occurs in cattle but much less frequently in field cases than in experimental ones. In one study involving beef cattle chronically infected with A. marginale in southern Brazil, a transplacental infection rate of 10.5% was obtained from 30 cows with no history of acute anaplasmosis during gestation.28 Abortion, neonatal infection, and fatal congenital infection have also been reported.29 In ewes intrauterine infection appears to occur with ease in experimental cases, provided the ewe is exposed during the latter two-thirds of pregnancy. Animal and Environmental Risk Factors Breed Bos indicus, Bos taurus, and their crosses have equal susceptibility to infection and show the same age susceptibility, but under field conditions B. indicus are not as commonly affected, probably because of their relative resistance to heavy tick infestation. However, the effects of the disease on body weight and clinicopathological parameters are the same for the two races of cattle. Breeds with black or red coat color have a higher risk of infection than those with white coats in regions where biting flies are the insect vectors. Dairy breeds may be at greater risk for iatrogenic transmission. Nutritional Status Clinical disease is less severe in cattle on a low plane of nutrition. Exposure of infected, clinically normal animals to devitalizing environmental influences, particularly shortage of feed, and the presence of other diseases may result in the development of acute anaplasmosis. For example, cattle introduced into feedlots are highly susceptible, and outbreaks among them are not uncommon 2 to 3 weeks after entry. Season In temperate climates a seasonal occurrence of disease occurs in association with seasonal occurrence of the insect vectors. Winter outbreaks are likely associated with iatrogenic transmission9 or possibly the winter tick, D. albipictus. Age at Infection All cattle are susceptible to infection, but age at infection is a major determinant of the severity of clinical disease. Young calves are less susceptible to infection with A. marginale than older cattle and, when infected, are

less susceptible to clinical disease. The reason for this is not understood, but splenectomized calves are fully susceptible to infection, which may be more severe than in the adult. Infection between 6 months and 3 years of age has increasing risk of clinical illness. Animals infected for the first time after 3 years of age are commonly affected by a peracute and fatal form of the disease. The age-specific incidence of clinical disease recorded in an outbreak in the United States showed 81% of cases in cattle aged between 2 and 4 years, with 94% of cases in cattle 3 years of age or older. Geographic Region Clinical disease is rare in enzootic areas because the infection pressure is high and cattle are infected at an age when they are age-resistant to clinical disease. The average age at which calves in enzootic areas become infected is 11 weeks (range of 4 to 24 weeks), and the clinical and hematological changes in them are mild and brief. Animals that have become seronegative for whatever reason in an infected environment are fully susceptible to infection. Clinical disease occurs where there is introduction of susceptible animals into endemic areas or the expansion of the vector population into previously free areas or into the interface between endemic and nonendemic regions. Case-fatality rates are usually high in outbreaks, but the mortality rate varies widely depending on susceptibility and may be 50% or more in cattle introduced to enzootic areas. Case-fatality rates of 29% to 49% are recorded in outbreaks in the United States; recovered animals are emaciated, and there is a prolonged convalescence. Pathogen Risk Factors Phylogenetic analysis of the MSP1a of A. marginale worldwide supports the existence of clades, and their evolution is linked to ecological traits affecting the tick vector performance.1 Consequently, some strains evolved under conditions that support pathogen biological transmission by R. microplus, whereas other strains may be linked to transmission by other tick species or to mechanical transmission in regions where R. microplus is currently eradicated.1 Australian isolates do not appear to differ significantly in antigenicity or virulence. In contrast, in other countries there can be significant differences between isolates in antigenic composition, the protection afforded against heterologous challenge, and virulence. It has been demonstrated that the phenomenon of infection exclusion occurs with A. marginale. Infection of tick cells and bovine erythrocytes with one genotype of A. marginale excludes infection with other genotypes. In herds of cattle from endemic areas where many genotypes are detected, only one genotype is found per animal.

771

Furthermore, cattle inoculated with two A. marginale isolates become infected with only one isolate. However, concurrent strain infections (superinfections) have been reported. For example, experimental infection with a low-pathogenic strain of A. marginale did not prevent infection with a highly pathogenic isolate; instead, it provided clinical protection against the highly pathogenic strain.30 Similarly, A. centrale–vaccinated cattle can be superinfected with field strains of A. marginale because they differ in their MSP2 genes.31 Furthermore, superinfection with A. marginale is associated with a significant increase in variant diversity, and high levels of endemicity also drive pathogen divergence toward greater strain diversity.32 Economic Importance Costs are from death and abortion in clinical cases, loss of production in sick and recovered animals, and costs associated with preventive measures such as tick control. There have been no recent estimates of cost, but anaplasmosis was estimated to cost $875 million in Latin American nations in 1977 and $300 million per year in the United States in 2003. In developed countries with the disease, exports of cattle to countries that do not have it are constrained. A major cost in developing countries is the constraint on efficient production and the limit to the introduction of susceptible cattle breeds with superior genetics. Experimental Reproduction Anaplasmosis can be reproduced experimental my subinoculating infected blood intravenously into intact or preferably splenectomized susceptible animals or by feeding infected ticks on them.

PATHOGENESIS

Anaplasma are obligate intraerythrocytic bacteria. They infect mature erythrocytes by an endocytic process that possibly involves the twin-arginine translocation (Tat) pathway, which exports fully folded proteins out of the cytoplasm of the bacteria with the type IV secretion system (T4SS).33–34 Inside the erythrocyte, bacterial reproduction occurs by binary fission to produce two to eight infective initial bodies that leave by exocytosis to infect other erythrocytes. The number of infected erythrocytes doubles every 24 to 48 hours, and the infection becomes patent 2 to 6 weeks after infection, with the time frame influenced by the initial challenge dose. Depending on the strain and the susceptibility of the host, from 10% to 90% of erythrocytes may be infected in the acute stage of the disease. At least 15% have to be infected before there is clinical disease. Infected erythrocytes are removed by phagocytosis in the reticular endothelial system, with release of acute-phase inflammatory reactants and the consequent development of fever.

772

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Continued erythrocyte destruction occurs, resulting in the development of mild to severe anemia and icterus without hemoglobinemia or hemoglobinuria. Anaplasmosis is primarily an anemia, with the degree of anemia varying with the proportion of infected erythrocytes. The first appearance of the bacteria in the blood coincides with a fall in the hematocrit and erythrocyte levels, the appearance of immature erythrocytes in blood smears, and the development of fever. Acutely affected animals may die shortly after this phase is reached. The appearance of anti-erythrocyte antibodies late in the acute stage may exacerbate the anemia. If the animal recovers from the initial acute attack, the disease goes into the subacute and chronic phase. The degree of anemia varies widely in young cattle up to 3 years of age but is always severe in adults and in splenectomized animals. Cattle that survive the disease become carriers for life and serve as reservoirs of A. marginale because they provide a source of infective blood for both mechanical and biological transmission by ticks. They have lifelong immunity and are resistant to clinical disease on challenge exposure. Carrier animals have cycles of parasitemia associated with the development of new antigenic variants to allow new cycles of invasion, multiplication, and destruction approximately every 10 to 14 days. In the carrier animal, the concentration of infected erythrocytes varies markedly at bimonthly intervals from 103 to 105 infected cells/mL of blood, much lower than in the acute phase.9 Each cycle reflects the emergence of one or more clones that express a unique, hypervariable region (HVR) of MSP2 and MSP3.35 These “escape variants” are not recognized by antibody present at the time of emergence. As the new variants replicate, a new variantspecific antibody controls them. This cycle continues unabated, allowing lifelong persistent A. marginale infection.5,36 It is believed that IgG-2 antibody is responsible for controlling the initial acute bacteremia and the subsequent peaks of bacteremia that arise during persistent infection, and that the antibody acts either by neutralizing extracellular bacteria in the process of invading new erythrocytes or by opsonizing bacteria that are then targeted for phagocytosis.37 The process is not sufficient to clear infection, and cattle remain persistently infected for life. Another factor that may contribute to persistence is the rapid deletion of antigenspecific CD4+ T lymphocytes following infection and the failure to establish a strong memory T-cell response during the course of the disease.38–39

CLINICAL FINDINGS Cattle There are few recent reports on clinical findings during outbreaks of bovine anaplasmosis. The incubation period varies with

the challenge dose but is generally several weeks with tick-borne infection and 1 to 5 weeks with the inoculation of blood. In most cases the disease is subacute, especially in young animals. Rectal temperature rises rather slowly and rarely to above 40.5° C (105° F). It may remain elevated or fluctuate with irregular periods of fever and normal temperature alternating for several days to 2 weeks. Anorexia is seldom complete. Death can occur at this stage, but many survive in an emaciated condition, and their fertility is impaired. The mucous membranes are jaundiced and show marked pallor, particularly after the acute stage is passed, but there is no hemoglobinuria. Peracute cases, with a sudden onset of high fever, anemia, icterus, severe dyspnea, and death, often within 24 hours, are not uncommon in adult dairy cows. Affected animals are often hyperexcitable and tend to attack attendants just before death. Pregnant cows frequently abort. In convalescent bulls there may be depressed testicular function for several months. In a recent outbreak in Hungary, two of five acutely affected cattle died, but the herd had concurrent infection with other pathogens.40 A fatal congenital infection in a 2-day-old calf was associated with bovine viral diarrhea virus infection in South Africa.29 In another case, clinical anaplasmosis was diagnosed in a 15-year-old cow, 13 years after eradication of the main vector Rhipicephalus microplus in Okinawa, Japan, implying that the cow was a long-time persistent carrier.41 Sheep and Goats In sheep and goats, infection is usually subclinical, but in some cases, particularly in goats, a severe anemia may occur, and a clinical picture similar to that found in cattle may be seen. Severe reactions of this type in goats are most frequent when the animals are suffering from concurrent disease. Goats may show hyperexcitability and may bite at inanimate objects. The experimental disease in lambs includes fever, constipation or diarrhea, pale and icteric conjunctivae, and severe anemia 15 to 20 days after inoculation. The anemia is not completely resolved in 3 to 4 months.

CLINICAL PATHOLOGY Hematology Anaplasmosis in cattle, sheep, and goats is characterized initially by normocytic normochromic anemia, which becomes macrocytic normochromic as the disease develops. Erythrocyte destruction may be so severe that the erythrocyte count is reduced to 1.5 million/µL. Immature red cells are common at this stage, and their presence is considered to be a favorable sign. The small dot-like bacteria are discernible at the periphery of up to 10% of the red cells in subacute cases, but in peracute cases more than 50% of the cells

may be infected. A. ovis are usually situated at the periphery of erythrocytes, but as many as 40% of infested cells may show submarginal organisms. Reticulocytosis and basophilic stippling are usually evident.42 Packed cell volume and red blood cell count were found to be the most informative parameters in the routine clinical practice for A. ovis infection in sheep.43 Diff-Quik staining of blood smears is as accurate as Giemsa in the detection of A. marginale and can be completed in 15 seconds compared with nearly an hour for Giemsa. There are no diagnostic clinical chemistry findings, but as evidence of oxidative stress, levels of erythrocyte lipid peroxidation (LPO) and plasma nitrate (NO) may be elevated during the acute phase.44 Concentrations of acute phase proteins (Hp, SAA, ceruloplasmin, and fibrinogen) may also be elevated.45

SEROLOGY

Several serologic tests have been employed for epidemiologic studies of bovine anaplasmosis, but the two currently preferred for identifying infected animals are the competitive ELISA and the card agglutination test.46 The competitive enzyme-linked immunosorbent assay (cELISA) is the most accurate serologic test currently available for anaplasmosis; it uses a monoclonal antibody specific for MSP5.9 However, it cannot differentiate between A. marginale and some other Anaplasma species, because they all possess the MSP5 antigen. A dot-ELISA with high sensitivity, specificity, and predictive value is also described and could be particularly applicable to field examinations. For further details, please see Aubry.9 The card agglutination test (CAT) examines serum or plasma for antibodies against A. marginale. It is cheap, quick, and sufficiently accurate to be used as a herd test. Currently, in most countries, the card agglutination and complement fixation tests are routinely available. The complement fixation test (CFT) used to be the standard test for the detection of carrier animals. It is satisfactory for use in cattle, goats, and sheep, but the antibody titer is highest during the active phase of the disease and sufficiently low in carrier animals to give a proportion of false-negative results. False-positive reactions can occur because of erythrocyte contamination of the A. marginale antigen and the presence of antibodies to erythrocytes in some cattle sera. A number of other tests have been developed. A capillary tube agglutination test of comparable efficiency is available, is more economical and faster than the CFT, and is particularly suited to testing in extensive field situations. An indirect fluorescent antibody test (IFAT) is also accurate and particularly suitable for testing blood that has been dried onto paper

Amyloidoses

VetBooks.ir

for passage through the mail. It is also an accurate test for selecting recently affected animals. A rapid lateral flow assay (LFA) has been developed that is a useful tool for screening cattle moving from an area with infection to a disease-free area.47 The assay can be carried out in 10 to 15 minutes, it requires no expensive equipment, and it is comparable to laboratory tests in its performance. Vaccinated animals may react to all of the serologic tests for periods of over 1 year. Molecular Methods Molecular methods can now be used to detect low levels of bacteremia and to differentiate A. marginale from other species.48 The PCR assay is more sensitive than other methods in detecting anaplasmosis, especially in latent infections.49 Nested PCR procedures showed bacteremia with a sensitivity of 50 infected erythrocytes per milliliter or as few as 10 copies of A. marginale.50–51 Furthermore, a multiplex PCR assay has been developed for the simultaneous detection of Theileria annulata, Babesia bovis, and A. marginale in cattle,52 and the assay was successfully used to detect A. marginale, Babesia bigemina, and B. bovis during an outbreak of tick-borne diseases in southern Brazil.53 PCR assay can also be used to detect infected ticks. Transmission to splenectomized animals is used to detect carriers and to assess the efficacy of treatment or vaccination. It has been the gold standard for the demonstration of A. marginale–free blood, but it is expensive and is now largely replaced by PCR in most countries.

NECROPSY FINDINGS

The most obvious findings are emaciation, pallor of the tissues, and thin, watery blood. There is mild jaundice, and the liver is enlarged and orange. The kidneys are congested, and there may be myocardial hemorrhages. The spleen is enlarged, with a soft pulp. The bone-marrow cavity may be reddened by increased hematopoietic tissue in acute cases, but there may be serous atrophy of marrow fat in chronic cases. Postmortem identification of A. marginale can be established by staining blood smears with Giemsa or with direct fluorescent stains. Peripheral blood is superior to organ smears. Brain smears are unsatisfactory. The technique is applicable to fetuses suspected of being aborted as a result of infection with Anaplasma spp. Nucleic acid–based tests may be used but are rarely needed for routine diagnosis at necropsy. Samples for Confirmation of Diagnosis • Clinical pathology—blood smears from cut surface of an ear (CYTO, FAT) • Histology—fixed spleen, liver, bone marrow

773

Imidocarb (5 mg/kg IM twice, 7 days apart) (R-2)

every 28 days or by chlortetracycline in the feed at 1.1 mg/kg BW daily. Elimination of infection is difficult but not impossible. A trial testing the ability of chlortetracycline therapy to eliminate the carrier state examined three doses (4.4 mg/ kg per day, 11 mg/kg per day, and 22 mg/kg per day) of oral chlortetracycline fed for 80 days.56 A negative reverse-transcription PCR assay result was confirmed in all treated groups within 49 days and by cELISA some additional 49 to 88 days later. Subinoculation of splenectomized steers confirmed chemosterilization with oral chlortetracycline.56

Enrofloxacin (12.5 mg/kg SC twice, 48 hours apart) (R-1)

CONTROL

DIFFERENTIAL DIAGNOSIS Other causes of hemolytic anemia

TREATMENT TREATMENT AND CONTROL Treatment Oxytetracycline (6 to 10 mg/kg IM daily for 3 days, or 20 mg/kg IM single dose) (R-1)

Control Killed A. marginale vaccine (2 doses 4 weeks apart, then a booster) (R-1) Live A. centrale vaccine (single dose, preferably in yearlings) (R-2) Live A. marginale vaccine (single dose, yearlings only) (R-3) Attenuated A. marginale vaccine (R-4) Cell-culture live A. marginale vaccine (R-2)

Treatment is with tetracyclines, imidocarb, or enrofloxacin. Treatment of clinical disease can be with oxytetracycline, 6 to 10 mg/kg BW daily for 3 days, or a single injection of long-acting oxytetracycline at a dose of 20 mg/kg intramuscularly (IM). The convalescent period is long, and animals remain persistently infected. Concurrent administration of estradiol cypionate (14.3 mg/kg BW IM) appears to improve the rate of recovery by reducing rickettsemia during treatment. Blood transfusions are indicated in animals with a PCV less than 15%. Rough handling must be avoided. Imidocarb (5 mg/kg BW IM twice, 7 days apart) is also an effective treatment for clinical cases but does not eliminate persistence.54 The drug is not approved for use in the United States and Europe because it is a suspected carcinogen. Enrofloxacin (12.5 mg/kg BW SC twice, 48 hours apart) is effective, but it also results in persistent infection. At 7.5 mg/kg BW it provided faster reduction of rickettsemia and PCV recuperation compared with long-acting oxytetracycline at 20 mg/kg BW.55 The risk for infection in the rest of the herd should be assessed and, if necessary, temporary or prolonged protection should be provided. Protection can be provided by tetracyclines or by vaccination. Temporary protection in the face of an exposure risk can be achieved with a single intramuscular injection at 20 mg/kg BW of long-acting tetracycline. The results generally are good except when the cattle are exposed to infection during the 14 days before the treatment. Prolonged protection can be achieved by intramuscular injection at 20 mg/kg BW of long-acting tetracycline

Methods for the control of anaplasmosis have not changed greatly over the past several decades. Basically, the control and prevention measures include (1) maintenance of Anaplasma-free herds through import and movement control, testing, and elimination of carrier cattle; (2) vector control; (3) prevention of iatrogenic transmission; (4) administration of antibiotics; and (5) preimmunization with live vaccines and immunization with killed vaccines.9 The eradication of anaplasmosis is not a practicable procedure in most countries at the present time because of the wide range of insects capable of carrying the disease, the long period of infectivity of carrier animals, and, in some areas, the presence of carriers in the wild animal population. In enzootic areas some benefit is derived from the control of ticks and other vectors, and weekly dipping in an acaricide has been used in tropical areas to control this and other tick-borne diseases. However, acaricide use has limited efficacy in reducing tick infestations and is often accompanied by serious drawbacks, including the selection of acaricide-resistant ticks, environmental contamination, and contamination of milk and meat products with drug residues. Consequently, the possibility of vector vaccines that target tick proteins is now being investigated.57 Such vaccines can reduce tick feeding and reproduction, and they also reduce the infection and transmission of pathogens from the tick to the vertebrate host. A recombinant trial vaccine against the tick-protective antigen subolesin has the potential not only to control tick infestation of cattle, but also infection with multiple pathogens such as A. marginale and Babesia bigemina.58–59 General Measures Prior serologic testing should prevent the introduction of the disease into herds by carrier animals. Attention should also be given to preventing iatrogenic transmission with instruments used for injections or surgical operations by disinfection after use on each animal. This is particularly important in feedlots where introduced groups are often subjected to multiple vaccinations and implantation at a time when their resistance

774

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

is lowered by transport and change of feed. On such occasions, iatrogenic transmission can be greatly reduced by single-use needles or needle-free administration systems for parenteral administration of vaccines or antimicrobials.27 Movement of Animals Naive animals that are to be introduced into an enzootic area should be vaccinated. Some advantage can be gained when introducing animals into an enzootic area by limiting the introductions to animals of less than 2 years of age and by bringing them in when the insect population is least numerous. Elimination of Carriers Elimination of carriers is feasible in regions that are subject to only periodic incursions of infection and that do not have endemic tick vectors. It can be achieved by serologic testing and culling of reactors or by treating them with oral oxytetracycline over a prolonged period of time as outlined earlier.56 Outbreaks If an outbreak does occur, affected animals should be treated vigorously as described previously and in-contact animals vaccinated and/or placed on a regimen of prolonged tetracycline protection. Subsequently, all exposed animals should be tested serologically and the reactors treated or preferably salvaged. Prolonged treatment regimens can be used to provide protection to cattle in seasonal risk periods of transmission. Chemotherapy Chemotherapy for control is more commonly used in the United States than in other areas of the world. It can be of value in feedlot cattle but is not applicable to range cattle. It is expensive and carries the risk of causing selection of resistant strains. Vaccination Vaccines for the control of anaplasmosis are either live or killed vaccines. Both types use A. marginale or A. centrale from infected bovine erythrocytes, and although both types induce protective immunity that reduces or prevents clinical disease, neither type prevents cattle from becoming persistently infected with A. marginale. Cattle that have recovered from acute infection or have been immunized with killed vaccines are solidly protected against challenge with the homologous strain but are only partially protected against challenge with heterologous strains. Most control programs in enzootic areas are based on increasing the resistance of the population by immunization. In any vaccination program, particular attention should be paid to the animals at high risk, particularly animals brought in from nonenzootic areas, those in surrounding similar areas to which infection may be spread by expansion

of the vector population under the influence of suitable climatic conditions, and animals within the area that are likely to be exposed to climatic or nutritional stress. Killed Vaccines Killed A. marginale are usually in an adjuvant vehicle. The vaccine requires two doses, 4 weeks apart, with the last dose given at least 2 weeks before the vector season. Subsequently, booster doses should be given 2 weeks before the next vector season. The vaccine does not prevent infection but does significantly reduce the severity of the disease. It does have the advantage over the other vaccines of having a relatively short postvaccination period when animals remain positive to serologic tests. The duration of the immunity is at least 5 months. There is a risk for neonatal isoerythrolysis. The risk can be reduced by vaccinating only empty cows and avoiding unnecessary booster injections. When this vaccine is used in the face of an outbreak, tetracyclines can also be given to provide temporary protection during the period of development of immunity; tetracyclines do not interfere with the development of this immunity. Preliminary reports of the efficacy of DNA vaccines are not encouraging. Live Vaccines A living A. centrale vaccine is used extensively in Australia, Africa, Israel, and Latin America, but not in the United States, and there is some reluctance to introduce it into areas where A. centrale does not already occur. Living A. centrale vaccine is prepared from the blood of infected splenectomized donor calves and is stored chilled or frozen. The vaccine causes a mild, inapparent disease, but it can cause severe reactions in occasional animals. It is generally safe in young cattle. A single vaccination is used in endemic areas, and the immunity is reinforced by continuous challenge and considered to persist for life in tick areas. A. centrale and A. marginale share immunodominant epitopes and have similar antigenic variation in major surface proteins, both of which play a role in the cross-immunity that occurs.60 The immunity induced by the live A. centrale vaccine in cattle does not prevent subsequent infection with A. marginale by the infection exclusion phenomenon because both differ in their MSP2 genes.31 The efficacy of this vaccine varies geographically. Vaccination with A. centrale reduces the severity of the reaction when infection with A. marginale occurs but does not give absolute protection. Protection against challenge in Australia is adequate in most cases, and certainly sufficiently effective enough to justify its use. In contrast, the use of the same vaccine in countries other than Australia, where there are more antigenically diverse and highly virulent strains,

is often inadequate, and better vaccines are required. Tetracyclines will prevent establishment of infection and immunity by the vaccine and should not be administered for 3 weeks before vaccination. Living A. marginale has been used as a vaccine but its administration is limited to the relatively resistant age group below 1 year of age, to the winter months when vectors are sufficiently rare to avoid the chance of spread to other age groups, and to circumstances where animals that react severely can be restrained and treated adequately. The method has the serious disadvantage of creating a large population of carrier animals that may subsequently spread the disease. Attenuated vaccines have been attempted by irradiating strains, by passage of the organism through sheep or deer, and by the use of naturally low-virulence isolates. Although most have been received with initial enthusiasm, some have proved ineffective, and others have been associated with adverse reactions. Some are effective against strains in some geographic regions but give unsatisfactory protection against clinical disease in other regions. Problems With Live Vaccines All vaccines currently must be produced in live animals, which is expensive. With noninactivated vaccines, there is a risk of transmitting blood-borne viruses. In Australia, a single calf infected with bovine leukosis virus was unsuspectingly used in the production of A. centrale vaccine. The contaminated vaccine was given to 22,627 cattle in 111 herds and resulted in a high rate of infection with bovine leukosis virus in the vaccinated cattle. A cell-culture system has been developed for propagation of A. marginale in a continuous tick cell line derived from embryonic Ixodes scapularis. Recently, protective immunity was induced by immunization with a live, cultured A. marginale strain.61 Vaccinated calves had a stable PCV and low bacteremia following challenge with a virulent strain, whereas A. centrale only afforded partial clinical protection. Important features of this cell-culture candidate vaccine are that it carries no risk of biological transmission, it can be easily distinguished from field strains, and only one dose is required. Studies are ongoing for the development of safe and effective subunit vaccine containing epitopes critical to effective immunity. Whereas blood-derived wholeouter-membrane (OM) preparations and cross-linked surface proteins provide the best protection from high-level bacteremia and anemia,62 they may not be practical for large-scale production. Recombinant proteins, DNA vaccines, and killed preparations of A. marginale, including inactivated cellculture-derived organisms, have failed to

Amyloidoses

VetBooks.ir

recapitulate the protection seen with OMbased vaccines.61 The ideal vaccine for anaplasmosis in cattle (as well as in sheep and goats) would be one that prevents infection, induces protective immunity, and possibly blocks biological transmission from the tick to the vertebrate host.9 However, further research is needed to achieve this goal. FURTHER READING Aubry P, Geale DW. A review of bovine anaplasmosis. Transbound Emerg Dis. 2011;58:1. Brown WC. Adaptive immunity to Anaplasma pathogens and immune dysregulation: implications for bacterial persistence. Comp Immunol Microbiol Infect Dis. 2012;35:241. Howden KJ, et al. An update on bovine anaplasmosis (Anaplasma marginale) in Canada. Can Vet J. 2010;51:837. Kocan KM, et al. Advances toward understanding the molecular biology of the Anaplasma-tick interface. Front Biosci. 2008;13:7032. Kocan KM, et al. Antigens and alternatives for control of Anaplasma marginale infection in cattle. Clin Microbiol Rev. 2003;16:698. Kocan KM, et al. The natural history of Anaplasma marginale. Vet Parasitol. 2010;167:95. Merino O, et al. Tick vaccines and the control of tick-borne pathogens. Front Cell Infect Microbiol. 2013;3:30. OIE manual of diagnostic tests and vaccines for terrestrial animals; World Organisation for Animal Health 2012; chapter 2.4.1:589. Passos LM. In vitro cultivation of Anaplasma marginale and A. phagocytophilum in tick cell lines: a review. Rev Bras Parasitol Vet. 2012;21:8.

REFERENCES

1. Estrada-Pena A, et al. BMC Biol. 2009;7:57. 2. Kocan KM, et al. Vet Parasitol. 2010;167:95. 3. Cabezas-Cruz A, et al. PLoS ONE. 2013;8:e65243. 4. de la Fuente J, et al. Vet Microbiol. 2007;119:382. 5. Chavez AS, et al. PLoS ONE. 2012;7:e36012. 6. Zivkovic Z, et al. BMC Vet Res. 2007;3:32. 7. de la Fuente J, et al. Vet Microbiol. 2007;119:375. 8. Haigh JC, et al. J Wildl Dis. 2008;44:569. 9. Aubry P, Geale DW. Transbound Emerg Dis. 2011;58:1. 10. Silveira JA, et al. Transbound Emerg Dis. 2012;59:353. 11. Rahman WA, et al. Trop Biomed. 2012;29:66. 12. Tembue AA, et al. Rev Bras Parasitol Vet. 2011;20:318. 13. Ashraf QU, et al. Ticks Tick Borne Dis. 2013;4:395. 14. Coetzee JF, et al. Can Vet J. 2010;51:862. 15. Pare J, et al. Can Vet J. 2012;53:949. 16. Carelli G, et al. Ann NY Acad Sci. 2008;1149:107. 17. Kubeiova M, et al. Parasite. 2012;19:417. 18. Hornok S, et al. Vet Microbiol. 2007;122:316. 19. Lobanov VA, et al. Transbound Emerg Dis. 2012;59:233. 20. Boes KM, et al. Vet Clin Pathol. 2012;41:77. 21. de la Fuente J, et al. Res Vet Sci. 2008;84:382. 22. Hornok S, et al. Vector Borne Zoonotic Dis. 2011;11:1319. 23. Baldridge GD, et al. J Med Entomol. 2009;46:635. 24. Hornok S, et al. Vet Parasitol. 2010;174:355. 25. Da Silva AS, et al. J Parasitol. 2013;99:546. 26. Scoles GA, et al. J Med Entomol. 2008;45:109. 27. Reibold JB, et al. Am J Vet Res. 2010;7:1178. 28. Grau HA, et al. Rev Bras Parasitol Vet. 2013;22:189. 29. Pypers AR, et al. J S Afr Vet Assoc. 2011;82:179. 30. Bastos CV, et al. Vet J. 2010;186:374.

31. Molad T, et al. Vet Microbiol. 2010;143:277. 32. Ueti MW, et al. Infect Immun. 2012;80:2354. 33. Nunez PA, et al. PLoS ONE. 2012;7:e33605. 34. Lockwood S, et al. PLoS ONE. 2011;6:e27724. 35. Futse JE, et al. Infect Immun. 2009;77:3181. 36. Palmer GH, et al. NY Acad Sci. 2006;1078:15. 37. Brown WC. Comp Immunol Microbiol Infect Dis. 2012;35:241. 38. Han S, et al. J Immunol. 2008;181:7759. 39. Han S, et al. Clin Vaccine Immunol. 2010;17:1881. 40. Hornok S, et al. Res Vet Sci. 2012;92:30. 41. Ooshiro M, et al. Vet Parasitol. 2009;160:351. 42. Yasini S, et al. Iran J Parasitol. 2012;7:91-98. 43. Ciani E, et al. Acta Vet Scand. 2013;55:71. 44. De United Kingdom, et al. Trop Anim Health Prod. 2012;44:385. 45. Nazifi S, et al. Vet Microbiol. 2012;155:267. 46. OIE manual of diagnostic tests and vaccines for terrestrial animals. 2012; chapter 2.4.1:589. 47. Nielsen K, et al. J Immunoassay Immunochem. 2009;30:313. 48. Carelli G, et al. Vet Microbiol. 2007;124:107. 49. Ashuma, et al. Asian Pac J Trop Med. 2013;6:139. 50. Molad T, et al. Vet Microbiol. 2006;113:55. 51. Decaro N, et al. J Vet Diagn Invest. 2008;20:606. 52. Bilgic HB, et al. Exp Parasitol. 2013;133:222. 53. Canever MF, et al. Korean J Parasitol. 2014;52:507. 54. Coetzee JF, et al. Vet Ther. 2006;7:347. 55. Facury-Filho EJ, et al. Rev Bras Parasitol Vet. 2012;21:32. 56. Reinbold JB, et al. Vet Microbiol. 2010;145:69. 57. Merino O, et al. Front Cell Infect Microbiol. 2013;3:30. 58. Almazan C, et al. Vaccine. 2012;30:265. 59. Merino O, et al. Vaccine. 2011;29:8575. 60. Agnes T, et al. Infect Immun. 2011;79:1311. 61. Hammac GK, et al. Vaccine. 2013;31:3617. 62. Noh SM, et al. Infect Immun. 2008;76:2219.

EQUINE GRANULOCYTIC ANAPLASMOSIS (EQUINE GRANULOCYTIC EHRLICHIOSIS, ANAPLASMA PHAGOCYTOPHILUM) Anaplasma phagocytophilum causes disease of horses, humans, dogs, cattle, cats, and other mammalian species. It is characterized in horses by fever, depression, limb edema, icterus, and ataxia.1 The disease is described here with emphasis on that occurring in horses. See the next section, “Tick-Borne Fever,” for a description of the disease in other species.2

ETIOLOGY

The disease in horses is associated with infection by A. phagocytophilum, the same agent that causes human granulocytic ehrlichiosis (HGE). The organisms Ehrlichia equi, E. phagocytophilum, and the HGE agent are now classified as A. phagocytophilum.3 The variety of species affected and geographic distribution of the disease suggests strains of A. phagocytophilum of varying pathogenicity and host specificity. A. phagocytophilum is a recognized cause of tick-borne fever in sheep, goats, cattle, horses, dogs, cats, roe deer, reindeer, and humans in Europe and elsewhere.4,5 Infection of some domestic and wild ruminants, including deer, does not

775

induce clinical signs, which could be a result of host susceptibility, the strain of the organism, or combinations of both. There are multiple strains of A. phagocytophilum, and each has a distinct host tropism (reservoir host and vector), with disease being associated with infection of nonhost species (dogs, horses, and humans) by pathogenic strains of the organism.6–8 Strains of A. phagocytophilum that cause disease in dogs and horses in southern Sweden differ slightly in their genetic composition from isolates derived from North America. Similarly, nucleotide sequences of strains of A. phagocytophilum from the West Coast of the United States differ from those of strains originating from the East Coast. Strains of A. phagocytophilum in the western United States varying in their pathogenicity in horses, with strains isolated from horses with the disease (and from chipmunks and tree squirrels) producing severe disease in horses, whereas a strain derived from woodrats does not cause disease in horses.6,8 There is genetic variability in strains capable of causing disease in horses demonstrated in Germany and the Czech Republic.9,10 The organism can be isolated from lizards, which are believed to be the host in a reptile–tick–reptile cycle. A. phagocytophilum is an obligate intracellular bacterium that replicates in cells derived from the bone marrow (granulocytes).

EPIDEMIOLOGY Distribution The disease in horses occurs in the Americas (the United States, including California, Washington, Oregon, Minnesota, Wisconsin, and the southeastern and the northeastern states; and Brazil), France, Italy, Switzerland, Sweden, Germany, Poland, the Czech Republic, the Netherlands, and the United Kingdom.9,11–14 The prevalence of horses with serum antibodies to E. equi (A. phagocytophilum) in endemic areas of California is 10%, compared with 3% in areas where the disease is uncommon. On farms where the disease occurs frequently, 50% of horses have serum antibodies to E. equi (A. phagocytophilum). Approximately 18% of horses in areas of the upper Midwest of the United State in which Ixodes spp. ticks are endemic have antibodies to E. equi (A. phagocytophilum), whereas 4 % of horses in areas in which the tick does not occur are seropositive. In a convenience sample of 96 horses in the Czech Republic, 73% were seropositive (indirect fluorescent antibody testing).15 A survey of 563 horses in Lazio region of Italy (near Rome), where the disease in horses occurs, revealed a seroprevalence of 0.3%, whereas 41 of 300 (13%) of horses from Latium, Umbria, and Marche in central Italy were seropositive by immunofluorescent antibody testing for antibodies to A. phagocytophilum, and 20 (6%) were PCR positive.14

776

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

There is extensive evidence of exposure of dogs to A. phagocytophilum and of disease consistent with granulocytic ehrlichiosis;16 47% of dogs in endemic areas in California have antibodies to E. equi (A. phagocytophilum), and some show clinical signs consistent with the disease. There is evidence of widespread infection of other species with the organism; 0.2% of 2725 serum samples from cattle in California had detectable antibodies to A. phagocytophilum, and 43% to 96% of deer and moose, respectively, in Norway are seropositive. Serologic evidence of infection is widespread in Europe.4 However, demonstration of antibodies in serum by surveys does not provide information on the pathogenicity of the infecting strains of A. phagocytophilum (which can vary widely in their capacity to produce disease). Anaplasma and Ixodes Ecology A. phagocytophilum is maintained by a cycle of infection of particular host species, including wild cervids or, in apparently separate cycles, small mammals such as the whitefooted mouse and dusky-footed woodrat in the United States and hedgehogs in Europe, and specific vector species of ticks.6,7,17–19 The tick–host–tick sylvatic cycles are now being identified, and there appears to be multiple such cycles, with more than one cycle (i.e., different host) being possible in a geographic area. Infection of horses, dogs, and humans, which are not part of the natural cycle of the organism, occurs through the bite of A. phagocytophilum–infected ticks. The organism is transmitted by hard ticks that are members of the Ixodes persulcatus complex, which includes Ixodes pacificus, Ixodes scapularis, and Ixodes ricinus. Transstadial, but not transovarial, transmission occurs. The tick vectors of A. phagocytophilum pass through four stages in their life cycle: egg, larva, nymph, and adult. Maturation from larva to nymph, maturation from nymph to adult, and egg laying all require the ingestion of a blood meal. Because transovarial transmission of infection does not occur, larvae or uninfected nymphs become infected by feeding on an infected mammal. The engorged and infected immature tick then dismounts and matures to the next life stage away from a mammalian host. When the immature tick reaches the nymph or adult stage, it again seeks a mammalian host. Transmission of the infection from the tick to a mammal occurs through feeding of an infected nymph or adult on a susceptible host. Environmental factors that affect the type of host species, its density in a particular area, and the number and activity of ticks are likely important in determining risk of infection of nonhost species (e.g., horses). Changes in local vegetation, such as that produced by clearing of forest and subsequent regrowth, can influence host and vector pop-

ulation densities and hence risk of transmission of the agent to nonhost species.20 Animal Risk Factors Horses that have not been exposed to A. phagocytophilum are susceptible to infection and disease. There is a marked seasonality of the disease in California, with most cases occurring in late autumn, winter, and spring. This seasonality likely correlates with the well-documented changes in populations of various stages of the vector ticks and host mammals.18 Horses of any age are susceptible, and there is no apparent breed or sex predisposition. Infection in horses is followed by a solid immunity, and recovered animals are resistant to the disease for at least 20 months, although it is suggested that reinfection and disease can occur. Serum antibodies persist for at least 300 days after infection in some horses but decrease to low levels in most horses by 200 days after infection. Transmission As discussed previously, transmission is through the bite of an infected tick. Transmission through use of blood-contaminated veterinary equipment or by blood transfusion is possible, with the latter being used to induce disease in experimental challenges. Perinatal transmission of A. phagocytophilum is reported in humans. Morbidity and Mortality The case-fatality rate is low, approximately 4%, and deaths of horses with uncomplicated disease are rare. Zoonotic Potential There is no evidence that infection spreads directly from infected horses or dogs to humans. However, dogs have been suggested to be sentinel animals, in that humans in areas in which dogs have a high prevalence of antibodies in serum to A. phagocytophilum might be at increased risk of infection from bites of infected ticks.

PATHOGENESIS

Following experimental infection, horses have organism detectable by PCR beginning 5 days after infection, with development of fever and depression 7 to 8 days after infection. Inclusions in granulocytes are detectable beginning 9 days after infection, at which time there is edema of the limbs. The organism incites a proinflammatory cascade after infection; administration of dexamethasone, which inhibits this inflammatory cascade, diminishes the severity of the disease in experimentally infected horses.21 The disease in horses is associated with rapid changeover of expressed p44 genes such that there is marked antigenic variation in the major surface protein, p44, during infection in an animal. The rapid changeover

of expression of p44 is attributed to development of specific antibody to the hypervariable region of p44. Infection in sheep results in immune suppression secondary to granulocytic and lymphocytic leukopenia, impaired antibody production, reduced lymphocyte response to mitogens, and a decreased oxidative burst activity of neutrophils. The prominent clinical sign of edema is likely related to the vasculitis that is characteristic of the disease.

CLINICAL SIGNS

The incubation period for the spontaneous disease is less than 2 weeks. Subclinical infections are believed to be common, based on the number of horses with serologic evidence of infection but no history of disease. Clinically there is high fever of 40° to 42° C (104° to 107° F) followed by mucosal pallor, jaundice, anorexia, depression, increased respiratory movement, incoordination and reluctance to move, and, after 3 to 4 days, edema and heat of the extremities. There can be petechial hemorrhages on mucosal membranes and pleural or peritoneal effusion. Edema persists for 7 to 10 days, and clinical signs resolve in 14 days. Clinical disease is more severe in horses over 3 years of age and is minor in young horses. Severely affected horses can have signs consistent with neurologic disease, including ataxia, defects in conscious proprioception, and recumbency.22 Arrhythmias can occur during the acute phase of the disease. Chronic infection and disease is not recognized. Death of an experimentally infected horse within 2 days of development of clinical signs was associated with disseminated intravascular coagulation.23

CLINICAL PATHOLOGY

There is commonly mild anemia and leukopenia. Thrombocytopenia is common in the acute stage of the disease. There are no consistent serum biochemical abnormalities. Positive identification of the disease is made on the presence of inclusion bodies (morulae) in the cytoplasm of neutrophils and eosinophils. Careful and protracted microscopic examination of a blood smear, stained with Giemsa, may be necessary to identify the inclusions (morulae). The inclusions are apparent as pleomorphic bodies of a blue–gray color, often in a spoke-wheel formation, in the cytoplasm of granulocytes. The number of infected cells can be quite small, and examination of a buffy-coat preparation may increase the sensitivity of the test. Diagnosis is achieved through use of a PCR test to identify A. phagocytophilum DNA in blood samples of infected horses and by demonstration of an increase in antibody titer detected by indirect fluorescent antibody staining. However, antibody titers are low to undetectable in approximately 44% of horses at the onset of clinical signs, and they

Amyloidoses

VetBooks.ir

reach a maximum within 1 month of infection. An ELISA that detects antibodies against the p44 surface antigen of A. phagocytophilum is suitable for use in dogs and horses. Evaluation of one commercial version of this test did not support its clinical use.24

NECROPSY EXAMINATION

At necropsy there are petechiae and edema of the legs, and at histologic examination there is vasculitis. There are often inflammatory lesions in the brain, heart, and kidneys. DIFFERENTIAL DIAGNOSIS Differential diagnoses include the following: • Equine infectious anemia, which has a much more protracted course and does not respond to treatment • Purpura hemorrhagica, which is often associated with infectious upper respiratory tract disease • Liver disease • Viral encephalitis • Equine herpesvirus-1 myeloencephalopathy, • Rabies • Botulism • Equine viral arteritis

TREATMENT TREATMENT Treatment of equine granulocytic anaplasmosis: • Oxytetracycline 7 mg/kg q 12 to 24 hours for 5 to 7 days (R1)

The specific treatment is oxytetracycline (7 mg/kg BW IV, every 24 hours) for approximately 5 to 7 days. Penicillin, streptomycin, and chloromycetin are not effective. The response to treatment with oxytetracycline is rapid; the fever is reduced or eliminated in 12 to 24 hours, and signs of the disease resolve within 5 to 7 days in most horses. Inclusion bodies are difficult to find 24 hours after beginning treatment. Without treatment the disease is usually self-limiting to 2 to 3 weeks.

the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1464. 3. Pusterla N, et al. J Equine Vet Sci. 2013;33:493. 4. Stuen S. Vet Res Comm. 2007;31:79. 5. Dziegiel B, et al. J Med Micro. 2013;62:1891. 6. Foley JE, et al. Emerg Infect Dis. 2009;15:842. 7. Rejmanek D, et al. J Med Micro. 2012;61:204. 8. Foley J, et al. In: Sparagano OAE, et al., eds. Animal Biodiversity and Emerging Diseases: Prediction and Prevention. 2008:94. 9. Jahn P, et al. Vet Rec. 2010;166:646. 10. Silaghi C, et al. Parasite Vector. 2011;4. 11. Adaszek L, et al. Zoonoses Pub Hlth. 2011;58:514. 12. Burgess H, et al. Can Vet J. 2012;53:886. 13. Butler CM, et al. Vet Rec. 2008;162:216. 14. Laus F, et al. J Vet Med Sci. 2013;75:715. 15. Praskova I, et al. Ticks Tick Borne Dis. 2011;2:111. 16. Carrade DD, et al. J Vet Int Med. 2009;23:1129. 17. Morissette E, et al. Emerg Infect Dis. 2009;15:928. 18. Rejmanek D, et al. Ticks Tick Borne Dis. 2011;2:81. 19. Silaghi C, et al. Ticks Tick Borne Dis. 2012;3:49. 20. Foley JE, et al. Am J Trop Med Hyg. 2009;81:1132. 21. Davies RS, et al. Clin Vaccine Immunol. 2011;18:1962. 22. Gussmann K, et al. Schweiz Arch Tierheilkd. 2014;156:345. 23. Franzen P, et al. Vet Rec. 2007;160:122. 24. Veronesi F, et al. Vector Borne Zoonotic Dis. 2014;14:317.

EPERYTHROZOONOSIS SYNOPSIS Etiology Hemotrophic mycoplasmas (previously Eperythrozoon species) Epidemiology Subclinical infection common; clinical disease precipitated by stress. Horizontal transmission by blood. Vertical transmission important in swine. Clinical findings Acute icteroanemia or chronic ill-thrift in sheep and swine. Reproductive inefficiency and neonatal anemia in swine. Syndromes in cattle less defined. Clinical pathology Anemia and bilirubinemia. Blood smear for parasite in early disease. Serology useful as flock/herd test. Polymerase chain reaction (PCR). Treatment Tetracyclines, organic arsenicals. Control Nonspecific. Prophylactic administration of tetracyclines.

CONTROL

There is no vaccine, and specific control measures cannot be recommended at this time, although minimizing access of ticks to horses would appear prudent. There is no need to isolate infected horses. FURTHER READING Dziegiel B, Adaszek L, Kalinowski M, Winiarczyk S. Equine granulocytic anaplasmosis. Res Vet Sci. 2013;95:316-320. Pusterla N, Madigan JE. Equine granulocytic anaplasmosis. J Equine Vet Sci. 2013;33:493.

REFERENCES

1. Dziegiel B, et al. Res Vet Sci. 2013;95:316. 2. Radostits O, et al. Equine granulocytic anaplasmosis. In: Veterinary Medicine: A Textbook of

ETIOLOGY The disease is associated with hemotrophic mycoplasmas, formerly thought to be rickettsial parasites and classified as Eperythrozoon. They infect a range of mammalian species and cannot be grown in culture. Species in farm livestock that have been associated with disease are Mycoplasma (Eperythrozoon) ovis in sheep, M. suis in swine, and M. wenyonii in cattle. Additional forms have been identified in sheep and cattle, designated Candidatus Mycoplasma haemobovis and Candidatus Mycoplasma haemovis, respectively, and the taxonomy of

777

the hemotrophic mycoplasmas is a work in progress.

EPIDEMIOLOGY Occurrence Eperythrozoonosis occurs in sheep, swine, cattle, and llamas, with the greatest clinical occurrence and importance in swine and sheep. Latent eperythrozoonosis also occurs in several species of deer, elk, and goats. The organisms appear species specific, although M. ovis has been transmitted from sheep to goats, identified in a number of species of farmed and free-ranging deer,1,2 and detected by PCR of the blood of a Texas veterinarian.3 Three distinct species of hemotrophic mycoplasmas, including two novel species, have been identified by PCR in white-tailed deer in the United States.4,5 The distribution is as follows: • Sheep. Eperythrozoonosis of lambs associated with M. ovis is recorded in Africa, Iran, the United States, Canada, Great Britain, France, Norway, Germany, Poland, Eastern Europe, Australia, and New Zealand • Pigs. Also known as infectious anemia of pigs, the disease is recorded mainly in the United States, Canada, Great Britain, and continental Europe. • Cattle. Eperythrozoonosis in cattle is widely distributed, with reports from North and South America, Africa, Australasia, the British Isles, continental Europe, and the Middle East. • Llamas. Infection with Eperythrozoon spp. is reportedly widespread in llamas in the United States. Infection has been detected in animals that also had other disease problems or as the result of specific survey studies, and it is likely that the organism acts primarily as a secondary opportunistic pathogen in llamas. Source and Transmission The reservoir of infection is the persistently infected animal, and the disease can be transmitted by any mechanism that transfers infected blood. In sheep it is thought that the minimal infective dose is one parasitized erythrocyte. Horizontal and vertical transmission are possible. Sheep The method of natural spread of the infection in sheep is probably via biting insects. Serologic studies in Australian sheep show that the prevalence of farms with infection is high and that spatial differences are probably a result of differences in vector occurrence. In an infected flock or herd, the disease can also be spread by management practices that transfer infected blood. In sheep, these include vaccination, ear-tagging, shearing, and mulesing, although these risk factors have not been associated with infection in any epidemiologic studies.

778

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Pigs Skin parasites and blood-contaminated needles and instruments have been shown to transmit disease in swine. Transplacental transmission is also important in swine. Host and Pathogen Risk Factors Seasonal differences in disease prevalence occur, being more common in the summer and autumn. This corresponds to increased vector populations but also to weaning of lambs and management procedures that may encourage spread, such as the multiple use of needles for vaccination. Regional differences in the clinical severity of the disease has led to postulations of differences in virulence between strains of the organism. There may also be genetic difference in host susceptibility, and field observations are that the Merino is more susceptible to infection and disease. The increased scrutiny of isolates using PCR has identified novel strains of hemotrophic Mycoplasma in many animal species, some of which are yet to be confirmed as new species. Many studies suggest that subclinical infection is common and that the development of clinical disease requires the presence of some other debilitating factor or stress for disease to occur. In Merino sheep this may be the stress of weaning, suboptimal nutrition, and intercurrent gastrointestinal nematode infections. Viral infections with porcine reproductive and respiratory syndrome (PRRS) and swine influenza appear to predispose its occurrence in swine. Intercurrent infections of hemotrophic Mycoplasma with other blood parasites, such as Anaplasma, are recorded in association with severe disease in sheep and cattle.6,7 However, a phenomenon known as interference, in which infection with one blood parasite protects against another, has been noted with Theileria and hemotrophic Mycoplasma in cattle.8

PATHOGENESIS

Following experimental infection there is a variable prepatent period, usually 1 to 3 weeks, which is followed by a period of intense parasitemia. Ring-form, coccoid, and rod-shaped organisms are evident in stained blood smears. The organism is epicellular, infecting the surface and periphery of erythrocytes, and is also found free in the plasma in blood examinations.9 There is a profound hypoglycemia during the parasitemic phase, which is believed to be a result of direct consumption of glucose by the parasite. The period of intense parasitemia lasts for a period of 5 to 10 days, following which visible organisms in the blood become much less frequent and anemia develops. Parasitized erythrocytes are removed from the circulation by the spleen. It is believed that the parasite alters the erythrocyte membrane, exposing new antigenic determinants and stimulating the development of

antierythrocyte antibodies.9 The severity and duration of the anemia vary between individuals, but the disease commonly lasts from 1 to 2 months. Upon recovery there may be further cycles of parasitemia and anemia, which are less severe. Sheep that develop a high antibody titer tend to rapidly clear the Parasitemia, whereas sheep that have a poorer antibody response tend to show persistent parasitemia and recurrent episodes of anemia. Once an animal is infected, it is probably infected for life.

CLINICAL FINDINGS Sheep Sudden death and deaths associated with exercise, accompanied by hemoglobinuria and icterus, may be a feature in some sheep and some outbreaks, but, more commonly, the disease is manifest with fever and depression followed by the development of anemia, exercise intolerance, and ill-thrift. In some cases, it may be the principal cause of ill-thrift in lambs. There is reduced wool growth, and in the experimental disease in lambs at pasture, a retardation of growth of up to 2 kg has been recorded 5 weeks after infection. Lambs suckled by infected ewes are passively immunized via the colostrum until weaning. Pigs Acute icteroanemia is the classical syndrome and occurs in feeder pigs. It is characterized by weakness of the hind legs, mild fever (40° C; 104° F), increased pulse rate, pallor of the mucosae, and emaciation. Jaundice is a frequent but inconsistent feature of the disease. Case fatality is high, and death occurs 1 to 5 days after the onset of clinical signs. Although once quite common, the prevalence of this form has decreased, possibly as a result of the use of feed additives containing arsenicals and effective ectoparasite control. Another manifestation includes anemia and weakness in neonatal pigs accompanied by low piglet viability, affecting several litters. Affected pigs are pale and lethargic, and there is marked variation in birth weight within affected litters. Low-birth-weight piglets die shortly after birth. The anemia increases in severity between birth and weaning age, the pigs have skin pallor and exercise intolerance, and there is considerable variability in weaning weights. The syndrome may or may not be accompanied by reproductive inefficiency characterized by delayed estrus cycles and embryonic death. Anemia, jaundice, and poor growth rate can also present primarily in weaner pigs. Subclinical infections associated with subclinical anemia are reported to result in reproductive failure, anestrus and delayed estrus, reduced sow body condition, increased susceptibility to enteric and respiratory disease, and failure of feeder pigs to gain weight at the expected rate.

Cattle Clinical disease has been considered uncommon and has largely been a problem in cattle that have been splenectomized for experimental use, with disease occurring 1 to 4 weeks after the splenectomy. However, clinical disease is recorded in adult commercial dairy cattle manifest with lassitude, stiffness, pyrexia, diarrhea, hindlimb and udder edema, and a fall in milk production, with one reported case clustered 5 days on either side of vaccination against bluetongue virus.10 Eperythrozoonosis has also been associated with a syndrome occurring in heifers in early to midlactation, during late summer and early autumn, in which there was fever, swelling of the teats and the hindlimbs, lymph node enlargement, and a fall in milk production. Signs of infection resolved in 7 to 10 days, regardless of treatment. A similar transient disease occurring in the spring and summer months, and manifest with scrotal and hindlimb edema and infertility, has been associated with eperythrozoonosis in young bulls.

CLINICAL PATHOLOGY Blood Smears and Hematology The presence of the organism can be established by examination of a blood smear taken during a clinical episode and when the animal has fever. In countries where there is no serologic test available this may be the only method of diagnosis. Parasitemia is most intense before the development of clinical anemia and appears as 0.5- to 1.0-mm, coccoid, rod- or ring-shaped basophilic particles on red cells or free in plasma. Parasitemia is difficult to detect once clinical signs of disease are evident and very difficult in chronic disease. It is recommended that blood samples from a number of animals in the group be examined if eperythrozoonosis is suspected. Lowered values for hemoglobin and packed cell volume (PCV) are evident on hematological examination of clinically affected animals, and there is marked red cell anisocytosis and polychromasia with basophilic stippling and the presence of many Howell–Jolly bodies in sheep. A profound hypoglycemia may be demonstrated, and there are elevated concentrations of unconjugated and total bilirubin. Polymerase Chain Reaction The development of conventional and realtime PCR assays now offers a more precise, sensitive, and efficient diagnostic method, with many animals negative on blood smears being positive on the PCR test.10 Serology Sheep The complement fixation test and indirect fluorescent antibody test (IFA) have been used. With the complement fixation test, sera

Amyloidoses

VetBooks.ir

from affected animals give positive reactions on the third day of clinical illness, remain positive for 2 to 3 weeks, and then gradually revert to negative. Chronic carriers of the disease are usually negative reactors. The IFA or ELISA tests are more suitable for serologic studies because infected animals remain seropositive for significantly longer periods.

4. Boes KM, et al. Vet Clin Path. 2012;41:77. 5. Maggi RG, et al. Comp Immunol Microbiol Infect Dis. 2013;36:607. 6. Hornok S, et al. Vet Microbiol. 2009;136:372. 7. Hornok S, et al. Res Vet Sci. 2012;92:30. 8. Tagawa M, et al. Vet Parasitol. 2013;195:165. 9. Hoezle LE, et al. Vet J. 2014;202:20-25. 10. Strugnell BW, et al. Cattle Practice. 2011;19:75.

Pigs The indirect hemagglutination test and ELISA test can be used in swine and are of value in herd diagnosis but may not detect infection in an individual pig, especially those under 3 months of age. Experimental challenge of splenectomized piglets may be used to determine the presence of infection. PCR may resolve laboratory diagnostic problems.

BOVINE PETECHIAL FEVER (ONDIRI DISEASE)

TREATMENT

A single intramuscular injection of tetracycline or oxytetracycline (3 mg/kg BW or more) is an effective treatment in sheep, with clinical improvement occurring in 24 hours in the early stages of the disease. Chronic infections are less responsive. Treatment of affected lambs with neoarsphenamine (30 mg/kg BW) or Antimosan (6 mg/kg BW antimony) is effective in relieving clinical illness but does not completely eliminate the parasite. Imidocarb dipropionate also is effective in treatment, but recrudescence at 2 to 4 weeks is common.

CONTROL

Control of disease in sows and neonates has been reported with the inclusion of chlortetracycline in the sow feed at 300 g/ton or by intramuscular administration of oxytetracycline to sows at 14 and 7 days before the expected farrowing date. Tetracyclines can also be used in feed or by in-line water medication in feeder pigs. With large flocks of sheep in enzootic areas, reinfection or recrudescence occurs so quickly that control by treatment may be an unwarranted expenditure. In confined swine operations, the detection of carrier pigs by PCR, and their subsequent removal, has been proposed as a possible control procedure. FURTHER READING Hoezle LE. Haemotrophic mycoplasmas: recent advances in Mycoplasma suis. Vet Microbiol. 2008;130:215-226. Hoezle LE, et al. Pathobiology of Mycoplasma suis. Vet J. 2014;202:20-25. Radostits O, et al. Eperythrozoonosis. In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1154-1156. Strugnell B, McAuliffe L. Mycoplasma wenyonii infection in cattle. In Pract. 2012;34:146-154.

REFERENCES

1. Grazziotin AL, et al. J Wild Dis. 2011;47:1005. 2. Grazziotin AL, et al. Vet Microbiol. 2011;152:415. 3. Sykes JE, et al. J Clin Microbiol. 2010;48:3782.

Bovine petechial fever is a rickettsiosis of cattle caused by Ehrlichia (Cytoecetes) ondiri. The disease occurs in the highlands of Kenya and Tanzania at altitudes of 1500 to 3000 m, although it is considered likely to occur in neighboring countries with similar topography. Characteristically, bovine petechial fever occurs in cattle that break out from fenced pastures and graze adjacent forest or bushland areas, or when they are grazed on these areas at the end of the dry season. It also occurs in cattle that have been recently introduced to these areas, and indigenous cattle appear to acquire resistance. Epidemics occur in cattle imported to infected areas and last 1 to 2 months, involving 60% to 80% of the group and resulting in significant losses. However, no outbreak has been reported for over a decade. Infection can be experimentally transmitted to cattle, sheep, goats, wildebeest, impala, duiker, bushbuck, and other wild ruminants, but natural disease is seen only in cattle. E. ondiri infection is believed to be endemic in wild ruminants, especially bushbuck and duiker, and the disease sporadically spills into cattle grazing forest edges or scrubs. The vector is not known, although epidemiologic findings suggest a tick vector of restricted distribution. The disease in cattle is characterized by high fever and widespread petechial hemorrhages in mucous membranes for periods up to 10 days; epistaxis, melena, and unilateral conjunctivitis occur in more severely affected animals. The eyeball is tense, protruding through swollen, everted conjunctival sacs as the so-called “poached-egg eye.” Pregnant animals may abort, and there is a fall in milk production for several weeks in lactating animals. Anemia may be severe enough to result in death 3 to 4 weeks after infection. There is a profound lymphocytopenia by the second day of infection, followed by leukopenia and thrombocytopenia. The organism can be demonstrated in granulocytes and monocytes in Giemsa-stained blood smears during the febrile period, but it cannot yet be cultured. Serology (indirect fluorescent antibody test) can be carried out to detect antibodies against E. ondiri. Grossly, the main lesions of ondiri disease are widespread petechial hemorrhages and enlarged, congested lymph nodes. In severe cases, death is often a result of severe hemorrhages into the lungs and airways. Abomasal

779

mucosa is edematous, and the contents of the ileum and colon are tarry. Differential diagnosis includes other hemorrhagic diseases of cattle, such as acute trypanosomiasis, acute theileriosis, hemorrhagic septicemia, bracken fern poisoning, Rift valley fever, and heartwater. Tetracyclines are effective in treating early experimental cases but are ineffective in advanced cases. Recovered animals may be latently infected and are immune to reinfection for at least 2 years. Control is by avoiding grazing cattle in forest edges and in paddocks with patches of thick scrub. FURTHER READING Blowey RW, Weaver AD. Color Atlas of Diseases and Disorders of Cattle. 3rd ed. New York: Mosby/ Elsevier; 2011:237. Sumption KJ, Scott GR. Bovine petechial fever (Ondiri disease). In: Coetzer JAW, Tutsin RC, eds. Infectious Diseases of Livestock. Vol. 1. 2nd ed. Cape Town: Oxford University Press; 2004:536. Valli VEO. Bovine petechial fever. In: Maxie GM, ed. Pathology of Domestic Animals. Vol. 3. 5th ed. Philadelphia: Saunders/Elsevier; 2007:310.

MYCOPLASMA SUIS INFECTION IN PIGS Mycoplasma suis causes anemia, fever, and icterus in pigs and was formerly called Eperythrozoon sui; it is a member of the Mollicutes family. The classical disease is now called infectious anemia of pigs. The original descriptions of the condition had two possible causes, E. suis and E. parvum. Currently, on the basis of 16S rRNA, E. suis is classified as M. suis within the Mycoplasma genus. Pigs infected with E. parvum have only mild disease. In a recent study1 in Brazil, a novel species was discovered, and by PCR this showed 98% to 99% matched identity to Candidatus M. haemominutum and is likely to be E. parvum. This disease is probably greatly underdiagnosed because of the difficulties in diagnosis. A recent study in China showed a heavy infection rate in swine workers (32/65),2 and it is now being isolated from diseased humans. Outbreaks of hemotropic mycoplasma infection have been recorded in Mongolia.3 It can cause acute and chronic infections.

ETIOLOGY

M. suis is a rod-shaped, coccoid, or ring-like bacterium, about 0.2 to 2.0 um in diameter. It is seen attached to the surface of red blood cells (RBCs), but recent studies have shown that it can also invade RBCs and live in vacuoles or free in the cytoplasm.4 It has not yet been cultured, but the genome has been sequenced.5 It exists in two clusters, a North American/European form and a Chinese form,6 and these were also found in wild boar in Germany.7

780

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

EPIDEMIOLOGY

VetBooks.ir

The disease can be found in all ages of pigs. Maternally derived antibody may play a part in preventing or delaying infection. It is probably found worldwide in pigs, but few surveys have been carried out. It has been found in Brazil8 and has also been found in deer.9 In affected herds, it is usually widespread and may cycle in waves through the herd. It was found in 13.9% of feeder pigs, and 40.3% of pig farms were positive in Germany.10 The morbidity can be 10% to 60%, and the mortality can reach 90% in an acute outbreak in very young pigs. Transmission is via blood and blood components by licking, biting, cannibalism, urine discharges, and other biological components containing blood. It is probably transmitted by biting flies, mosquitoes, and hypodermic needles. Transplacental infection has been suggested but not proven. The carrier state probably also exists. Predisposing factors may include immune-modulating viruses such as porcine reproductive and respiratory syndrome (PRRS) virus, porcine circovirus type II (PCV-2), and possibly simian immunodeficiency virus (SIV). Other factors may include increased arthropod activity and periods of stress, such as farrowing and weaning.

PATHOGENESIS

Initially, there is a heavy bacteremia, with colonization of the surface of the RBCs aided by a surface protein of the M. suis. The RBCs are then recognized as abnormal by the spleen and removed from circulation, adding to the anemia. The degree of anemia is closely related to the load of M. suis on the RBCs. The bacteria are also capable of penetrating the cell by an unknown method,4 and this may be the means of their persistence as an infection because they can then evade host defense mechanisms, increase virulence, and reduce antimicrobial efficacy. It is thought that the M. suis uses the erythrocyte glucose for its own metabolism and by damaging the cell membrane induces production of auto-agglutinins favoring removal of these cells by the phagocyte system, thereby increasing extravascular hemolysis. Any immune response may occur not only as a result of the M. suis but also in response to the infected RBCs, thereby increasing the hemolysis and anemia and subsequent jaundice. The infection itself may affect T cells and cause immunosuppression. In many instances the immunosuppression caused by concurrent infections with PRRS, PCV-2, and SIV will also complicate the pathogenesis and clinical signs. The rate of destruction of RBCs may be so great that the demand for glucose by the M. suis outstrips the gluconeogenesis, and hypoglycaemia may result, especially in pigs of 0 to 7 days of age, which are already low in glycogen.11 Endothelial cell activation, widespread

endothelial damage, and adherence of RBCs to the endothelium is evident in M. suis infections.12 The suggestion is that M. suis has a tropism for endothelial cells, and this interferes with the protective function of the endothelium, resulting in hemorrhagic diatheses.

CLINICAL PATHOLOGY

In acute cases there is a peak bacteremia 7 to 14 days following infection, with bacteria in blood smears and variable types of anemia. Decreases in packed cell volume, total red cell count, and hemoglobin concentration occur because of massive red cell parasitism. Anemia and bilirubinemia results from the RBC destruction, with hypoglycaemia and acidosis sometimes occurring.

CLINICAL SIGNS

In natural acute cases, which are most common in postweaned pigs, there is lethargy, pyrexia, anorexia, variable icterus, cyanosis of the extremities, petechial bleeding and ecchymoses, and often death within several days. In chronic cases there is pyrexia; anorexia; reproductive failure; mastitis, metritis, and agalactia (MMA); reduced birth weights; weakness and anemia of neonates; poor growth rates; and ill-thrift, with a predisposition to secondary infections. In experimental splenectomized pigs the incubation period is 3 to 10 days. It is particularly acute in the pig under 7 days of age, in which the signs include pallor, fever, occasionally icterus, and cyanosis of the extremities, particularly the ears.2 More commonly there is a mild anemia and a reduced growth rate. There may be increased bleeding and in some cases navel bleeding in young pigs. The bleeding was shown to resolve with the use of tetracyclines, suggesting that M. suis may be associated with navel bleeding. Recovered pigs may show a reduced growth rate and unevenness in the litter. Skin hypersensitivity, pallor, and unthriftiness may be seen in the chronic infection. In sows, there may be fever, anorexia, lethargy, and agalactia, particularly around farrowing. A severe outbreak of dysgalactia was described in the United States starting 1 day after parturition and lasting 4 to 6 days.13

PATHOLOGY

In acute cases there is anemia, jaundice, splenomegaly, and serous effusions in the body cavities. In the chronic cases, secondary infections often mask any primary lesions from the M. suis.

DIAGNOSIS

The diagnosis is complicated because there is no culture available for the organism. It is therefore dependent on the clinical signs, the direct demonstration of the organism in blood smears (stained by Romanowska-type stains, Giemsa, or acridine orange) taken at

the time of collection of the blood samplem and the hematology results of anemia and bilirubinemia. The position has been improved by the development of PCR techniques. The first qPCR for M. suis was developed in Germany7 and was shown to be much more sensitive than the blood smear test.11 The blood test is entirely dependent on the number of organisms present. The test used on 120 samples from clinically normal, healthy pigs on 11 farms found M. suis on 6 farms and M. parvum on 18 farms, with 3 positive for both, using qPCR.8 Antibodies can be detected by ELISA tests. These tests have been developed using recombinant antigens,14 and initially these were of high sensitivity but low specificity. Because the antibodies may only last 2 to 3 months, t false negatives were common. The blocking ELISA described is more efficient.15 A new ELISA was shown to be 98.5% specific and 96.9% sensitive and much more efficient than an IHA test.16

TREATMENT

Usually a course of oxytetracycline at 20 to 30 mg/kg given daily parenterally will prevent the clinical signs. Pigs affected will not usually feed or drink properly, so administration in the feed is only effective as a prophylaxis. If given in the feed, it needs to be fed for a minimum of 14 days. Often it prevents anemia, but ir does not prevent outbreaks.

CONTROL

Usually on an infected farm an equilibrium is reached between the organism and the host, but it can be overcome by concurrent infections, bad management, and a poor environment, and thus these should be rectified. The complicating factors should be removed, such as secondary infections by vaccination (PRRS, SIV, PCV-2). Excessive use of vaccines should be avoided because they involve the use of needles. There is no specific vaccine. FURTHER READING Hoelze LE. Haemotrophic mycoplasmas—recent advances in Mycoplasma suis. Vet Microbiol. 2008;130:215-226.

REFERENCES

1. Biondo AW, et al. Rev Bras Parasitol Vet. 2009;18:1. 2. Yuan CL, et al. Am J Vet Res. 2009;70:890. 3. Hu Z, et al. Emerg Infect Dis. 2009;15:1139. 4. Groebel K, et al. Infect Immun. 2009;77:576. 5. Guimaraes AMS, et al. PLoS ONE. 2011;6:e19574. 6. Watanabe Y, et al. J Vet Sci. 2012;74:1315. 7. Hoelzle K, et al. Vet Microbiol. 2010;143:405. 8. Guimaraes AMS, et al. Vet Rec. 2007;160:50. 9. Watanabe Y, et al. J Vet Med Sci. 2010;72:1527. 10. Ritzmann M, et al. Vet Microbiol. 2009;133:84. 11. Hoelzle K, et al. J Microbiol Meth. 2007;70:346. 12. Sokoli A, et al. Vet Res. 2013;44:6. 13. Congli Y, et al. Vet Microbiol. 2010;142:303. 14. Hoelzle K, et al. Clin Vaccine Immunol. 2007;14:1616.

Amyloidoses

VetBooks.ir

15. Zhang CY, et al. Vet J. 2012;193:535. 16. Liu J, et al. Res Vet Sci. 2012;93:48.

EPIZOOTIC HEMORRHAGIC DISEASE (BLACKTONGUE) SYNOPSIS Etiology Epizootic hemorrhagic disease virus (EHDV), an arthropod-borne, doublestranded RNA virus of the family of Reoviridae, genus Orbivirus Epidemiology Infectious but noncontagious disease transmitted by biting midges. Wild ruminants, particularly white-tailed deer, develop severe clinical disease with high mortality. Historically, cattle were believed to be resistant to clinical disease, but recent outbreaks of EHD primarily affecting cattle have been reported. Small ruminants may seroconvert but do not develop clinical disease. Occurrence is dependent on the presence of competent vectors. In temperate regions the highest disease incidence is in late summer and fall; in tropical regions EHD occurs throughout the year. Clinical findings In white-tailed deer, rapid edema development on the neck and head; swelling of the tongue and conjunctivae. Hemorrhagic diathesis with bloody diarrhea, hematuria. and dehydration. More chronic presentation with erosions or ulceration in the buccal cavity. In cattle. sudden anorexia, decreased rumination, drop in milk production, weakness, short-term low fever, stiff gait. Morbidity rate dependent on infectious pressure in the herd; low mortality   rate. Necropsy findings In white-tailed deer. cyanosis and petechial ecchymotic hemorrhages of oral mucosa and tongue. Ulceration and necrosis of oral mucosa in more chronic cases. Pulmonary edema; serosanguinous fluid in thorax, pericardial sac. and abdomen. Rarely fatal   in cattle. Diagnostic confirmation Virus isolation and detection from blood or tissue (whole blood, spleen, lungs, lymph node,   liver). Serologic tests (agar gel immunodiffusion test [AGID], enzymelinked immunosorbent assay [ELISA], serum neutralization assay). Treatment Supportive treatment; no specific treatment is available. Control Culicoides vector control (insecticides, larvicides, insect repellents, management of culicoides breeding areas). Commercial vaccines are not available; autogenous inactivated vaccines have been used.

ETIOLOGY Epizootic hemorrhagic disease (EHD) virus is an arthropod-borne virus of the family of Reoviridae, genus Orbivirus, closely related

781

to bluetongue virus. At least seven different serotypes (serotype 1 through 8, with serotype 3 considered to be similar to serotype 1) exist, which have been regrouped in recent years.1 Some of these have antigenic relations with serotypes of bluetongue virus. The EHD virus (EHDV) can infect most domesticated and wild ruminants naturally; pigs are not susceptible to infection. Clinical disease is primarily observed in wild ruminants. White-tailed deer are most severely affected, followed by mule deer and pronghorn antelope. In recent years several disease outbreaks in cattle herds have been reported.

Host Occurrence Under natural conditions infection occurs in sheep, cattle, and wild ruminants. Natural infection does not appear to occur in goats. Historically, clinical disease was primarily observed in wild ruminants, whereas cattle and to a lesser extent sheep were considered as reservoir hosts for the virus. In recent years, outbreaks of EHD affecting cattle have been reported in different parts of the world. Among wild ruminants, white-tailed deer and to a lesser degree mule deer and pronghorn antelope are most susceptible to clinical disease.

EPIDEMIOLOGY

Method of Transmission The disease is not contagious and is almost exclusively transmitted biologically by specific species of Culicoides. Epidemiologic data suggest that Culicoides species transmitting EHDV are similar, although not necessarily identical, to species transmitting bluetongue virus (BTV), but the vector competence of involved Culicoides species may differ for both viruses. Possible differences in the environmental temperature and number of days at or above a specific environmental temperature required for effective virus replication within the vector for BTV and EHDV may have contributed to the differences in geographic progression of these two viruses observed in recent years in Europe.1 Culicoides breed in damp, wet areas, including streams, irrigation channels, muddy areas, and fecal runoff areas around farms. Habitats for them exist on the majority of farm environments. Only female Culicoides are hematophagous and feed on their main or preferred host species, requiring at least one blood meal for the completion of the ovarian cycle. They feed nocturnally on animals in open pens and fields, and the optimal temperatures for activity are between 13° (56° F) and 35° C (95° F). In temperate areas the disease is seasonal because Culicoides do not tolerate low ambient temperatures, resulting in a vector-free season during late fall and winter.

Occurrence Because EHD is a vector-borne viral disease, occurrence of infection depends on the presence of competent vectors. Clinical disease or serologic evidence for infection has been reported from North and South America, Africa, Asia, Australia, and, more recently, from the region surrounding the Mediterranean Basin, including Morocco, Algeria, Tunisia, Israel, Jordan, and Turkey.1 Although the geographic occurrence of EHD was similar to that of bluetongue during the last century, the recent northward progression of bluetongue from the Mediterranean Basin far into the European continent was not observed for EHD. Historically, two serotypes (EHDV-1, EHDV-2) were predominant among the wild ruminant population throughout the United States, except in the Northeast and the arid Southwest, and southern Canada. Between 2006 and 2009, EHDV-6 was recovered from clinical cases in Indiana, Illinois, Missouri, Kansas, Michigan, and Texas. In Australia, six serotypes (1, 2, 5, 6, 7, and 8) have been isolated, predominantly from sentinel cattle in the north. Three serotypes have been recognized in Africa: EHDV-3 (considered to be identical to EHDV-1), EHDV-4, and EHDV6.2 Two major outbreaks of EHD mainly affecting cattle that were associated with a genetically distinct strain of EHDV-2, called Ibaraki disease, occurred in Japan, Korea, and Taiwan in the 1960s and again 30 years later. The first epidemic of Ibaraki disease resulted in 39,000 sick cattle and 4000 deaths. In Europe and the Mediterranean Basin numerous outbreaks of EHD, primarily affecting cattle instead of wild ruminants, have been observed since the beginning of the millennium. In 2001 clinical cases associated with EHDV of unspecified serotype were reported in Israel. Between 2004 and 2007, outbreaks caused by EHDV-6 were reported in Morocco, Algeria, Tunisia, and Turkey. In 2006 an epidemic associated with EHDV-7 and affecting dairy and beef herds was reported in Israel.3 In 2008 EHD was added to the list of notifiable diseases of the World Organization of Animal Health (OIE) and is now considered an emerging disease in cattle.4

Culicoides Species Different Culicoides species have different geographic occurrence, and their distribution in a country is determined by climatic factors and the presence of a preferred host. In the United States C. sonorensis is the predominant vector throughout much of the country, except in the Southeast, where C. insignis predominates. C. imicola is a predominant vector; in the Middle East and Asia, C. imicola has been involved in the recent expansion of EHD, but C. obsoletus and C. pulicaris have been implicated as new vectors associated with recent EHD outbreaks.1 Other Vectors Other vectors may transmit the disease mechanically but are unlikely to be of major significance in disease epizootics.

782

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Other Methods of Transmission The Ibiraki strain (EHDV-2) was isolated from the internal organs of aborted fetuses, which proves that transplacentar virus transmission is possible. The incidence of congenital infection and thus the epidemiologic relevance of this route of virus transmission has not been quantified. No reports of isolation of EHDV in the semen of infected animals are available. Although oral and fecal shedding of EHDV-1 was reported in white-tailed deer, the epidemiologic importance of oro-fecal transmission has not been ascertained.1 Host Risk Factors Although most ruminant species are susceptible to infection with EHDV, clinical disease primarily is observed in white-tailed deer and, in recent years, also in cattle. Wild Ruminants In North America, EHD is considered one of the most important diseases of deer, particularly of white-tailed deer (Odocoileus virginianus) but also mule deer (O. hemionus) and pronghorn antelope (Antiocarpa americanna). There are areas of enzootic stability, where seroprevalance in deer is high but the clinical disease rare, and areas with low seroprevalance where clinical disease is severe. Cattle Historically, cattle were believed to function as reservoir hosts for EHDV by being susceptible to infection but resistant to clinical disease. The first reports of clinical disease related to EHDV in cattle date from 1959, when an epizootic disease called Ibaraki disease occurred in Japan. Disease outbreaks caused by the Ibaraki strain (EHDV-2) were subsequently observed in other Asian countries. Since 2001 a number of disease outbreaks have occurred in the Mediterranean Basin and also in the United States that have been linked to EHDV serotypes 6 and 7.5 EHD is now considered an emerging disease of cattle.4 Morbidity rates in cattle herds can be considerable, depending on the infectious pressure, but clinical signs are much less severe than in white-tailed deer, and mortality is low.5 Small Ruminants Sheep are susceptible to infection with EHDV but do not develop clinical disease. Although this species has been incriminated as a potential reservoir host for EHDV, more recent experimental studies do not support the assumption that sheep may play a relevant role in the epidemiology of EHDV.6 In goats, thus far only the presence of EHDV antibodies but not the presence of virus or viral DNA has been documented. It is therefore assumed that goats are resistant to infection.

Morbidity and Case Fatality Wild Ruminants Morbidity and case-fatality rates of EHDV in wild ruminants are difficult to determine. Among the Cervidae, white-tailed deer are the most severely affected, and case-fatality rates in this species are much higher than those in other commonly affected species, such as mule deer, black-tailed deer, and pronghorn antelopes. In the Northeast and Midwest of the United States EHD typically recurs every year in late summer and early fall, but mortality and case-fatality rates can vary greatly from year to year. Morbidity and mortality rates may be as high as 90%, although in most instances cases are mild and mortality is low. This variability is thought to be caused by a number of factors, such as the abundance of biological vectors, the ambient temperature, the serotype of EHDV that is circulating, herd immunity (based of previous exposure to a similar strain), and the genetic variation in the susceptibility of the host.7 Cattle Outbreaks of EHD in cattle herds have been reported in Asia, the United States, and the Mediterranean Basin. Although in rare instances considerable morbidity and casefatality rates have been reported, as in the case of Ibaraki disease outbreaks in Japan and Korea, in most cases morbidity rates do not exceed 5%, and deaths are uncommon. During an EHDV outbreak in Israel, withinherd morbidity rates between 5% and 80% were observed in dairy herds, but the mortality rate remained below 1%.3 Economic Importance Until outbreaks of EHD affecting cattle occurred, EHD was considered a disease primarily affecting wild ruminants and thus of minor economic importance. Following the most recent outbreaks in North Africa, Turkey, and Israel, the economic importance of EHD had to be reconsidered. Since 2008 EHD has been listed as reportable disease by the OIE, and EHD is now considered a potentially emerging disease of cattle.4 Because the increased virulence of EHDV for cattle has only been observed in recent years, not many studies investigating the economic impact of this disease are available. Losses result from decreased productivity, increased involuntary cull rates, abortion, and, in some instances, death.5 In infected herds in Israel EHD was found to cause an average loss in production of 125 kg/cow per year, an effect that was highly dependent on the season of the year the outbreak occurred and the seroprevalence of the herd. The highest losses were observed when outbreaks occurred in September and in herds with the highest seroprevelences.5 With losses related to increased mortality added to the production losses, the total cost for the dairy industry in Israel was estimated to range between

US$ 1,600,000 and 3,400,000, which is equivalent to an average loss of US$ 26.50 per cow.5

PATHOGENESIS

The incubation period for EHD in deer is 5 to 10 days. After infection with EHDV, initial viral replication occurs in endothelial cells of the lymphatic vessels and lymph nodes draining the site of infection.1 A viremic phase ensues during which the virus is disseminated to other sites of virus replication, such as the lymph nodes and spleen, causing secondary infection of the endothelial cells of arterioles, capillaries, and venules throughout the body. In blood, EHDV is associated with erythrocytes and to a lesser extent with lymphocytes. This cell association permits prolonged viremic periods and the concomitant presence of virus and antibodies in blood. Endothelial damage results in leakage of blood vessels and ensuing disseminated intravascular coagulopathy. Fibrin thrombi occluding small blood vessels cause congestion, hemorrhage, and edema of surrounding tissue. Ischemic necrosis occurs in tissue where blood perfusion is interrupted as a result of thrombosis. Occurrence of viremia in relation to EHDV infection was studied in cattle and deer. The virus could be detected from 2 days following infection, and all animals were viremic 4 days after infection. After experimental infection virus could be isolated up to 28 days following infection and in rare instances up to 50 days following infection.1 Neutralizing antibodies can be detected from 10 to 14 days following infection but do not prevent or interrupt viremia. The concomitant presence of virus and homologous antibodies is commonly observed in EHD during the first weeks following infection. Maternal antibodies were identified in fawns born to dams infected with EHD for up to 18 weeks. Passive immunity was not able to prevent infection or viremia in fawns exposed to EHDV but alleviated clinical signs of the disease.1

CLINICAL FINDINGS Deer In deer, EHD may occur in a peracute, acute, or chronic form. Peracute disease is characterized by high fever, anorexia, respiratory distress, and severe and rapidly developing edema of the neck and head. Conjunctivae and the tongue are commonly swollen. Affected animals may be found dead or die within 2 days of the first clinical signs.7 The acute form is the classical presentation of EHD in deer, in which extensive hemorrhage affecting the skin, gastrointestinal tract, and heart is the hallmark sign. There is often hyperemia of the conjunctivae and mucosal membranes of the mouth. Ulcers and erosions may develop on the tongue, dental pad, palate, rumen, and

Amyloidoses

VetBooks.ir

abomasum. Excessive salivation and nasal discharge, which can be tinged with blood, have been observed. Bloody diarrhea, hematuria, and dehydration can occur. The mortality rate for both the peracute and acute forms is generally high. Deer with the chronic form of EHD are sick for several weeks but recover gradually. Cracks in the hooves resulting from the interrupted horn growth resulting from the disease may cause lameness. Chronic lesions of the mucosa of the rumen, with erosions, ulcers, and scarification, can cause emaciation.7

hamster kidney cells (BHKs), calf pulmonary artery endothelium cells (CPAEs), or African green monkey kidney cells (Vero). The cytopathic effect produced by EHDV is only observed on cell lines of mammalian origin and becomes apparent between 2 and 7 days after inoculation. Virus identification from cell cultures is based on a serum neutralization or plaque inhibition test using reference antisera. Virus isolation is the most reliable confirmation of EHDV infection because there are difficulties with the interpretation of serologic test results. However, traditional isolation methods require 2 to 4 weeks.

Cattle Most infections in cattle are unapparent. Occasional disease associated with infection with EHDV is recorded in cattle in the late summer in the United States and has been reported in recent outbreaks in the region around the Mediterranean Basin. Common clinical signs are anorexia, reduced rumination, weakness, decreased milk production, stiff gait, and short episodes of pyrexia.6 Reddening and swelling of the oral mucosa with necrotic ulceration of the dental pad and behind the incisor teeth, cracking and sloughing of the skin of the muzzle, and hyperemia of the skin of the teats and udder may occur in some cases. Ibaraki disease is characterized by fever, hyperemia and edema of the mucosae, hemorrhages, ulcerative stomatitis with laryngeal and pharyngeal paralysis, salivation, and dysphagia. At postmortem there were hemorrhages in the pharynx and esophagus, and animals commonly died with aspiration pneumonia. Infection of pregnant cattle with Ibiraki virus can also result in abortion and stillbirths, and currently this seems a more common clinical manifestation. Fetuses infected between 70 and 120 days of gestation may develop hydroancephaly.7

Detection of Antigen or Nucleic Acid Immunohistochemical tests, including immunofluorescence, or molecular techniques such as in situ nucleic acid hybridization, reverse-transcription polymerase chain reaction (RT-PCR), or the dot blot test can be used for rapid sensitive and specific detection of antigen. Use of RT-PCR has proliferated because of its simplicity, rapidity, reliability, reproducibility, sensitivity, and specificity. However, tests that detect viral RNA prove exposure to the virus but do not necessarily indicate that infectious virus is still present. An antigen-ELISA and sandwich-ELISA have also been used for EHDV identification but are less sensitive than the PCR.

CLINICAL PATHOLOGY

Specific diagnosis is either by isolation of the virus, detection of viral antigen or nucleic acid, or detection of specific antibodies in serum. Serologic assays can detect prior exposure to EHDV but cannot establish if the animal is viremic and thus infectious. Serologic tests may be of limited values in regions where EHDV is endemic and seroconversion is common in the affected population. Materials that can be used for virus isolation include heparin or EDTA in the blood, biopsies, or postmortem tissue samples of the spleen, lung, lymph nodes, or liver. Virus Isolation Virus isolation commonly is carried out by tissue culture or culture in embryonated chicken eggs (ECEs). Cell lines used for this purpose can be of insect origin, such as the KC cell lines derived from Culicoides sonorensis, or mammalian cell lines, such as baby

Serologic Tests A number of serologic tests for detection of either group-reactive antibodies or serotypespecific antibodies are available. The commonly available tests include the complement fixation test (CFT), the agar gel immunodiffusion test (AGID), a number of different ELISA tests, and serum neutralization (SN). The AGID test is easy to perform and inexpensive but is also relatively insensitive and detects cross-reacting antibodies to other orbiviruses, such as bluetongue virus. Over the last decades the CFT and AGID have been replaced in many laboratories by the more rapid, sensitive, and specific competitive ELISA. A number of ELISA tests have been developed that use group-specific monoclonal antibodies, and these present valuable alternatives to the AGID for routine diagnosis and international trade. The competitive ELISA (cELISA), which is the most sensitive and highly specific group-specific test, is the preferred test for serodiagnosis of EHD. The serum neutralization test (SNT) is serotype specific and thus allows for differentiation between antibodies against specific EHDV serotypes. The biological detection system (either ECEs or cell cultures) is reacted with a reference serum for specific EHDV serotypes, and the amount of virus neutralization is determined. Although the SNT is highly sensitive and specific, it is also expensive and time-consuming and is therefore not used as a routine diagnostic procedure.

783

NECROPSY FINDINGS Deer Lesions in white-tailed deer and related species are similar to those described for bluetongue in sheep and cattle. In acute cases, there may be cyanosis of the oral mucosa and tongue, along with widespread petechial to ecchymotic hemorrhages. In more chronic cases, there may be ulcers and necrotic debris in the oral mucosa and severe hoof lesions, including fissures and sloughing. Lesions in yaks include swollen conjunctivae, ulcerated dental pads, mucoid sanguineous nasal discharge, and petechial hemorrhages in multiple organs; others are pulmonary edema and serosanguinous fluid in the thorax, abdomen, and pericardial sac.8 Cattle The disease is usually subclinical and nonfatal in cattle.9 Samples for Confirmation of Diagnosis • Histology—fixed oral and mucocutaneous lesions, abomasum, pulmonary artery, skeletal muscle from a variety of sites, left ventricular papillary muscle, brain from aborted fetus (LM, IHC) • Virology—chilled lung, spleen, CNS tissues, thoracic fluid from aborted fetus (ISO, PCR, in situ HYBRID, ELISA, etc.) DIFFERENTIAL DIAGNOSIS Foot-and-mouth disease Bluetongue (wild ruminants) Bovine viral diarrhea/mucosal disease (cattle) Malignant catarrhal fever (cattle) Bovine ephemeral fever (cattle)

TREATMENT There is currently no specific treatment available for EHDV.

CONTROL Reduction of Infection Through Vector Abatement Attempts to control EHD through a reduction of infection consist of reducing the risk of exposure to infected Culicoides and reduction in Culicoides numbers. Neither are particularly effective. Widespread spraying for Culicoides control is not usually practical and has only a short-term effect. To address recent EHD outbreaks in cattle herds, several affected countries have introduced control measures such as monitoring of wildlife reservoirs, quarantine, vector control programs on farms, and awareness campaigns for veterinarians and farmers.1 Vaccination There is no commercially available vaccine against EHD. Autogenous inactivated

784

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

vaccines produced from virus isolates from ill or diseased herdmates have been used in the United States to tackle EHD outbreaks in captive wildlife deer. In Japan, both a modified live and an inactivated vaccine derived from the Ibaraki-2 strain have been developed to control Ibaraki disease. This vaccine, which is administered once during the lowvector season, was found to be safe and effective in controlling the disease.1 FURTHER READING

Savini G, et al. Epizootic haemorragic disease. Res Vet Sci. 2011;91:1-17.

REFERENCES

1. Savini G, et al. Res Vet Sci. 2011;91:1-17. 2. Allison AB, et al. J Gen Virol. 2010;91:430-439. 3. Yadin H, et al. Vet Rec. 2008;162:53-56. 4. OIE. 2009 At: ; Accessed 15.11.2013. 5. Kedmi M, et al. J Dairy Sci. 2010;93:2486-2495. 6. Kedmi M, et al. Vet Microbiol. 2011;148:408-412. 7. Center for Food Security and Public Health. 2006 At: ; Accessed 15.11.2013. 8. Van Campen H, et al. J Vet Diagn Invest. 2013;25:443-446. 9. Breard E, et al. Res Vet Sci. 2013;95:794-798.

BOVINE IMMUNODEFICIENCYLIKE VIRUS ETIOLOGY

Bovine immunodeficiency-like virus (BIV), also known as bovine lentivirus-1, is a lentivirus, within the larger family of Retroviridae. The virus shares structural and genomic similarities with other lentiviruses, such as equine infectious anemia virus, caprine arthritis-encephalitis virus, maedi-visna virus, and the feline, simian, and human immunodeficiency viruses. BIV was first described in cattle in the United States in 1972. These viruses replicate primarily in the cells of the host’s immune system following their insertion as provirus into the genome of these target cells, thus establishing a chronic, lifelong infection. The lentiviruses are usually associated with specific diseases. However, a clear involvement of BIV in the development of a clinical syndrome is not well established.

EPIDEMIOLOGY Prevalence of Infection Seroepidemiological evidence indicates that BIV infection has a worldwide distribution. Seropositive cattle have been identified in the United States, the Netherlands, New Zealand, Australia, Bali, Indonesia, Brazil, and Canada, with estimates ranging from 1% to 5% of cattle being infected. In Italy, the prevalence is 5.8% in dairy cattle and 2.5% in beef cattle. In individual herds the prevalence of infection may be much higher. In the United Kingdom, the seroprevalence was found to

be 5.9% in dairy cattle and 5.0% in beef cattle. The dairy and beef herd prevalence rates were 60% and 59%, respectively. Although the prevalence of BIV infection in the United Kingdom is low, it is widespread. Recent studies, using DNA derived from semen and buffy-coat samples, analyzed by nested PCR, found no evidence of BIV infection in western Canadian cattle. A seroprevalence of greater than 50% was present in a dairy herd at a university in the southeastern United States, which is an area with a high prevalence of infection in the cattle population. There is some evidence that in some cattle herds with a high incidence of unthrifty animals, the prevalence of seropositive animals may be as high as 95%. The prevalence of BIV infection among dairy cattle in Ontario is low and may be associated with an economically important decrease in milk production. Cases of dual infection with BIV and BLV have been reported in Mississippi dairy cattle. The virus has been found in the seminal leukocytes of 82% of randomly selected semen samples from a bovine stud semen repository, suggesting the possibility that artificial insemination of dairy cows may have a major role in the transmission of the virus. BIV may be involved in the pathogenesis of mastitis in cattle as a result of its immunosuppressive effects, but no clear evidence is available. Retroviruses are heat labile and readily inactivated at 56° C (133° F), and pasteurization of milk for human consumption should provide an adequate safeguard. Feeding milk seeded with the virus and pasteurized before inoculation into calves is effective in inactivating the virus and preventing transmission. There is no evidence that the virus is a potential human pathogen. Methods of Transmission The virus is strongly cell-associated and may be transmitted with infected blood, colostrum, and milk that contains lymphoreticular cells. There is some evidence of transplacental infection of the virus in cattle. In dairy cows naturally infected with BIV and seropositive at parturition, 40% gave birth to calves that were BIV seropositive before receiving colostrum, whereas seronegative cows did not. Calves born with anti-BIVspecific antibody do not demonstrate increased risk of clinical disease during the neonatal period, but the calves born to dams that are seropositive at parturition appear to be at increased risk of occurrence of some clinical signs. The prevalence rate of infection among bulls housed in stud farms was 9.6% using serology and 12.6% using PCR for the presence of BIV provirus in peripheral blood leukocytes. BIV has no obvious morphologic effects on the embryonic development of cattle, and it is possible to obtain embryos at the transferable stage free of the virus from cows

infected with the virus. It is unlikely that BIV is associated with embryos with the zona pellucida intact derived by in vitro fertilization from oocysts obtained from infected animals or with oocysts fertilized with infected semen when embryos are washed as recommended by the International Embryo Transfer Society. Embryos from donors infected with the virus are not likely to transmit the virus to recipients and the resulting offspring.

PATHOGENESIS

The pathogenetic mechanisms of BIV infection are unclear. Its pathogenicity is controversial. It is uncertain if the virus is a primary pathogen or a primary immunodeficiency virus that predisposes the animal to secondary infections. Despite extensive experimental studies, the pathogenic significance of the virus is uncertain. Infection of cattle with BIV is associated with lymphoproliferation, lymphadenopathy, immunosuppression, neuropathy, and progressive emaciation. The virus was initially isolated from a cow with persistent lymphocytosis, lymphadenopathy, neuropathy, and progressive emaciation. However, overt clinical disease in seropositive cattle is rare, and experimentally induced infection in calves has resulted in only mild clinical consequences. Early studies of inoculation of calves with the virus resulted in lymphoproliferative disease, lymphocytosis, and persistence of the virus. Later studies have failed to reproduce significant clinical disease, which may in part be a result of the long incubation period. It is also possible that the lentiviruses have variable virulence because genetic variation produces viruses with both antigenic and biological heterogenicity in pathogenesis. Experimental infection of an 11-month-old calf with the virus was followed by the development of a T-cell lymphosarcoma, and the bovine leukosis virus was not present. The virus and its DNA have been detected in the blood and semen of experimentally infected bulls. However, the virus has not been detected in the semen, blood leukocytes, or semen leukocytes of samples supplied by artificial insemination centers. Retroviruses, including the lentiviruses, are characterized by the expression of the unique enzyme, reverse transcriptase, that facilitates the transcription of the RNA of an infectious virus to a complementary DNA copy. The viral DNA has the ability to become incorporated into the host’s cell nucleus as a “provirus.” Proviruses are noninfectious, can remain latent for many years, and persist in the presence of antibody. A change in the virus from its latent form to an infectious RNA virus can occur and depends on activation of the latently infected cells. The stimuli for activation can include concurrent

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

infection and stress, or both. Although other lentiviruses such as equine infectious anemia virus can cause severe clinical disease, the cause-and-effect relationship between BIV infection in cattle and clinical disease has not yet been documented.

CLINICAL FINDINGS

In the United States, naturally occurring BIV infection in Holstein dairy cattle in Louisiana has been described. Progressive weight loss was common, and concurrent infections included metritis, subcutaneous abscesses, purulent arthritis, laminitis, and infectious pododermatitis, fascioliasis, and mastitis. Reduced vitality, dullness, and stupor were also common. The course of the disease varied from 3 to 40 weeks.

CLINICAL PATHOLOGY

Detection of Virus.  A PCR test has been used to detect BIV in the blood and milk of BIV-seropositive cows. The virus can be detected in experimentally infected calves using PCR in peripheral blood mononuclear cells. Serologic Tests.  With the BIV ELISA, naturally occurring cases in dairy cattle are serologically positive. An indirect immunofluorescent antibody test has been used to detect seroconversion in experimentally infected bulls by 17 days after infection. The sensitivity and specificity of the indirect fluorescent-antibody assay (IFA) and the nested-set PCR have been compared using Bayesian techniques. The PCR is the more sensitive assay.

NECROPSY FINDINGS

Moderate to marked enlargement of hemal lymph nodes has been described. Lymphoid depletion is common and characterized by an absence of follicular development in nodes draining regions with secondary infections. Encephalitis characterized by meningeal, perivascular, and parenchymal infiltration with lymphocytes, plasma cells, and macrophages with perivascular edema has been observed. Several secondary infections have been observed in cattle with BIV infection, but the role of BIV as a predisposing pathogen is uncertain. FURTHER READING

Gonda MA. Bovine immunodeficiency virus. AIDS. 1992;6:759-776.

Enzootic Bovine Leukosis (Bovine Lymphosarcoma) SYNOPSIS Etiology Bovine leukemia virus (BLV), the causative agent of enzootic bovine leukosis

(EBL), is an exogenous C-type oncovirus in the Retroviridae family. Epidemiology Infection is widespread in all continents, although several countries have successfully implemented BLV eradication programs. Prevalence of infection varies between countries. Persistent aleukemic (AL) infection is most common, followed   by infection with persistent lymphocytosis (PL) in 30% of infected animals. Less than 5% of infected animals develop lymphosarcoma, the only clinically apparent form of EBL. Clinical disease is most common in mature cattle. Infected animals are the only source of the virus, which is transmitted horizontally by transmission of infected lymphocytes from parturition, contaminated surgical instruments, rectal palpation, and blood-sucking insects. Congenital infection in 4% to 8% of calves born to infected cows. Genetic makeup of animal determines risk of developing PL or lymphosarcoma. Economic losses as a result of loss in milk production traits, premature culling, carcass condemnation, and restrictions of international commerce. EBL is currently not considered a zoonosis. Signs No clinical signs during stage of AL and PL. Lymphosarcoma characterized by loss of body weight, inappetence, pallor, weakness, and loss of milk production. Enlargement of several or all lymph nodes. Abomasal ulceration. Congestive heart failure. Paresis and paralysis as a result of neural involvement. Stertor as a result of enlargement of retropharyngeal lymph nodes. Eventually weak and recumbent. Clinical pathology Serology for BLV virus using enzyme-linked immunosorbent assay (ELISA) or agar gel immunodiffusion (AGID). Detect virus by polymerase chain reaction (PCR) or sheep bioassay. Lesions Multicentric lymphoid tumors affecting all body systems, especially heart, digestive tract, nervous system, reproductive tract. Diagnostic confirmation Serology and detection of virus by PCR. Differential diagnosis list Sporadic bovine leukosis (SBL) Congestive heart failure as a result of traumatic pericarditis Lymphadenitis as a result of tuberculosis and actinobacillosis Compression of spinal cord Fat necrosis Tuberculosis Treatment None. Control Test and slaughter seropositive animals in herds and areas with low prevalence of infection. Use bulk-tank milk ELISA as screening test. Establish virus-free herds and certify by retesting. Control disease in herds and countries with high

785

prevalence of infection by limiting spread within herds, segregating positive animals, and preventing the introduction of infected animals.

ETIOLOGY The causative agent enzootic bovine leukosis is bovine leukemia virus (BLV), an exogenous C-type oncovirus in the Retroviridae family that is highly homologous to human T-cell lymphotropic virus 1 and 2. Infection occurs by transfer of infected lymphocytes from one individual to another and is followed by a permanent antibody response and, less frequently, development of persistent lymphocytosis (PL) or lymphosarcoma. It has leukemogenic activity, can be grown in tissue culture, and produces specific antibodies in calves and sheep.

EPIDEMIOLOGY Prevalence of Infection Leukosis in cattle was originally described in Germany in 1871. Reports of the disease in cattle became common following World War II, and most countries that raise cattle have reported the occurrence of the disease. The main presumed routes of transmission having led to this epizootic during the first part of the last century in Europe were close animal contact and the use of whole blood vaccination, a procedure used at that time to protect cattle from developing babesiosis.1 For this purpose young cattle susceptible to babesiosis were injected with 2 to 3 mL of citrated blood drawn from donor cows with a history of babesiosis before going on pasture. Live animal transports across the Atlantic Ocean brought BLV to the Americas, where it primarily spread by close contact between infected and susceptible animals. The infection is now common in cattle in Canada, the United States, and many countries in eastern Europe and South America and some Asiatic and Middle Eastern countries. Today large parts of Europe and New Zealand are officially free of EBL after successful implementation of EBL-eradication programs.2 In Australia the National Dairy Enzootic Bovine Leucosis Eradication Program (NDEBLEP) was established in 2008 and eradicated BLV from dairy cattle by 2012. The prevalence of BLV infection in adult beef cattle in Australia is assumed to be very low. In the United States a serologic survey conducted in 2007 reported a herd prevalence of 83.9% of BLV seropositivity among dairy herds.3 A similar census from 1996 revealed a within-herd prevalence of 25% or higher in BLV-positive herds in the United States.4 Recent epidemiologic surveys from different Canadian provinces reported herd prevalence rates reaching up to 89% and individual animal prevalence rates between 20.8% and 37.4%.5,6 In Argentina a marked increase of the prevalence was reported for dairy herds in the last decades.7

786

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

The individual animal seroprevalence was estimated with 33%, and the percentage of infected herds with one or more infected animals was 84%. The seroprevalence of BLV infection in breeding beef bulls under 2 years of age offered for sale in Kansas was 8.5%. This indicates that young bulls purchased for entry into recipient herds could be infected with the virus. The infection occurs in water buffalo in Brazil and in draught animals in Cambodia. An outbreak of enzootic bovine leukosis in Egypt was associated with the importation of Holstein–Friesian heifers and bulls from Minnesota in 1989 to form a closed dairy herd in upper Egypt. In 1996 clinical evidence of EBL occurred, and ELISA testing revealed a BLV seroprevalence of 37.7% in cattle under 2 years of age and 72.8% in animals over 2 years of age. Occurrence of Clinical Disease The occurrence of clinical lymphosarcoma in countries where the infection occurs has been estimated to be 1 per 1000 per annum; it has been estimated at 1 per 50,000 per annum in infection-free countries. Even in countries or areas where the infection and the disease are common, there are many herds that remain uninfected. Dairy cattle are much more commonly infected than beef cattle, and they have a much higher incidence of lymphosarcoma. In severely affected dairy herds, an annual mortality rate of 2% is unremarkable, and it may be as high as 5%. All breeds of cattle are susceptible to BLV infection. It occurs rarely in animals less than 2 years of age and increases in incidence with increasing age. The prevalence of infection is higher in large herds than in smaller herds.3 The higher prevalence in dairy herds compared with beef herds is probably a result of their closer confinement and the higher average age of the herds. There are a number of forms taken by the disease, as follows: • Aleukemic enzootic bovine leukosis (AL) infection • Enzootic bovine leukosis with persistent lymphocytosis (PL) • Enzootic bovine leukosis with tumors— the common form in adults Methods of Transmission Direct Contact Horizontal transmission is the usual method by which the virus is spread under natural conditions. It appears that close physical contact and exchange of contaminated biological materials are required for transmission. The virus is present mostly in lymphocytes and can be found in blood, milk, and tumor masses. Most susceptible cattle become infected by exposure to infected lymphocytes, and not by cell-free virus. Either 10 µl (45,240 lymphocytes) or 1 µl (4524 lymphocytes) of whole blood from a BLV-seropositive cow when injected into

calves resulted in infection and seroconversion. It is likely that a threshold number of approximately 100 BLV-infected cells is required to establish infection in the recipient. Therefore any means by which BLVinfected lymphocytes can be transmitted from one cow to another is a potential means of transmission. Natural transmission occurs mostly in cattle more than 1.5 years of age with an apparently increased risk of infection during the periparturient period and after entering the milking herd.7 This suggests that vaginal secretions, exudates and placentas from cows, and contaminated calving instruments may serve as sources of infected blood cells. A considerable number of newborn calves were found to contract BLV infection around parturition or during the first hours and days of life.7,8 Intrauterine infection occurs in 4% to 8% of calves born from BLVseropositive cows in naturally infected herds. These cases probably occur as a result of transplacental exposure to the virus during gestation. The virus has been found in the nasal secretions of infected cattle for 2 to 4 years, but there is no evidence that transmission to other animals occurred. Transmission experiments suggest that the virus is not present in saliva, but it does appear intermittently in urine. It is present in nasal and tracheal washings but only in cells, not as a free virus. Semen, Artificial Insemination, and Embryo Technology Most workers have failed to find the virus in semen and artificial insemination (AI). However, the virus has been found in semen collected by massage of the donor’s urethra and accessory glands per rectum, a procedure that is associated with contamination of semen with blood. Although transmission by AI has not been demonstrated, it is possible that semen containing infected lymphocytes transmission could serve as a source of the virus. Thus bulls at AI centers will be required to be serologically negative for BLV virus. Properly collected semen from BLV-seropositive bulls will not contribute to dissemination of viral infection. More recent studies found that natural service breeding of heifers, and to a lesser extent, cows, was associated with an increase in BLV prevalence.9 Fertilized embryos from donors infected with BLV have been transferred without infection of the fetus. It is possible to produce transferable-stage in vitro fertilized embryos that are free of the integrated BLV provirus, from oocytes that had been exposed to BLV during maturation. Iatrogenic Transmission Transmission can occur via infected blood that contaminates surgical instruments, such as dehorning gouges, ear tattooing pliers, and hypodermic needles used on infected and then susceptible animals without

disinfection. Transmission can also occur during blood transfusions and vaccines containing blood, such as those for babesiosis and anaplasmosis. Amounts of blood as small as 0.1 µL are capable of transmitting the infection. Thus the infection can be transmitted via the tuberculin intradermal test. However, although some studies have found that use of common needles for blood sampling of infected and noninfected cows at the same time poses a great risk of transmission of the virus to noninfected cows, other studies suggest that the quantities of infective blood passed during injection with common needles is too small to induce infection. The routine practices of brucellosis vaccination, ear-tagging, and tattooing in dairy herds did not seem to be associated with the spread of the disease, but infection could be reduced from 80% to 4% in heifers between the time of weaning to calving by altering dehorning methods. Transmission via infective milk is possible by the passage of infected lymphocytes through intestinal mucosal epithelium during the first few hours of life. However, the risk of infection via this route appears to be greatly reduced because of the presence of maternal antibodies in colostrum and milk.8 Rectal Palpation The virus can be transmitted by rectal inoculation of infected blood into cattle and sheep. Using blood-contaminated sleeves from palpating seropositive heifers to palpate seronegative cows resulted in transmission of infection, as evidenced by antibody formation. This poses the possibility that the virus can be transmitted by rectal examination of cattle, particularly in dairy herds, when a single rectal palpation sleeve is used repeatedly during reproductive tract examinations. Field studies examining the use of the same sleeve for more than one animal or an individual sleeve for each animal indicate that rectal transmission is a potential route of spread of BLV, but that it is related to frequency of palpation and age of cattle. Controlled studies of rectal palpation of cows in a dairy herd over a period of 22 months, using a single sleeve per animal or not changing the sleeve between an infected animal and seronegative animals, resulted in a 2.8fold increase in the risk of BLV infection. Thus rectal examination without a change of sleeve may be a risk factor in some herds. Insects Blood-sucking insects may be involved in transmission of the virus. Evidence implicating arthropod vectors in BLV transmission is indirect, involving experiments in which virus-carrying arthropods or parts of them were transferred to uninfected cattle. In several experiments, infected tabanids, other biting flies, and ticks were placed by hand on cattle and sheep. Minced mouthparts or hematophagous insects previously fed on

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

BLV-infected cattle also were injected into hosts. In some countries there is empirical evidence that the incidence of seroconversion is higher after the tabanid fly season. A space–time study found a significant positive geographic correlation between the rate of incidence of BLV infection and the density of the horse-fly population. Seasonal variations in the incidence rates also occur; the highest rates are generally observed during summer and the lowest during winter, spring, and early summer. There is also a time link between the rate of seroconversion and the variations in activity of the horse-fly population. Experimentally, the virus has been transmitted by horse flies, Tabanus fuscicostatus, from a seropositive cow to recipient calves and goats. Horse flies take relatively large blood meals, have a painful bite, and are often interrupted in feeding and must finish feeding on other animals. This behavior, the large number of flies, and the low volume of blood and small number of lymphocytes required to transmit BLV make tabanid flies candidates for mechanical vectors of the virus. The stable fly, Stomoxys calcitrans, has an insufficient mouthpart volume to carry enough blood lymphocytes to transmit the virus. Congenital Infection Intrauterine infection has been estimated to occur in 4% to 8% of calves born from BLVseropositive cows in naturally infected herds and thus is considered to play a minor role in the epidemiology of EBL. Infection of the fetus probably occurs as a result of transplacental exposure to the virus during gestation. Calves born from seropositive cows acquire colostral antibodies if they ingest colostrum, which appears to have a protective effect against BLV infection during the first days of life.8 Antibody levels decline during the first 6 to 7 months of life. In one study the minimum and maximum duration of colostral antibodies were 14 and 147 days, respectively, with a half-life of 36 days. The decay of colostral antibodies and the age at which a calf can be expected to become seronegative is a function of the quantity of BLV antibodies absorbed by the calf and the infection status of the calf. Interspecies Transmission Cattle are the only species infected naturally, although sheep and goats can be infected experimentally. The infection does not spread from cattle to commingled sheep or between experimentally infected and noninfected sheep. However, horizontal transmission of a naturally occurring lymphosarcoma in sheep is associated with an antigenically similar virus to the BLV. It is assumed that horizontal spread of the BLV from cattle to sheep does not occur. The experimental transfer of infection from cattle to sheep is effected so readily that it has become a preferred technique for testing for the presence of a virus.

Source of Infection In cattle, infection with the virus is permanent, spontaneous recovery has not been documented, and the proviral DNA is maintained in infected lymphocytes. The virus is located in lymphocytes initially in a covert nonproductive state, resulting in an inability of antibodies to arrest the infection, and multiplication of the virus is not necessary for survival or transmission. The virus is also capable of periodic antigenic change and circumventing control by immune mechanisms; thus, the infected animal remains a source of infection for life, regardless of the simultaneous presence of specific antibodies. This virus–host system is the same as that of other retroviruses, especially equine infectious anemia (EIA) and maedi-visna of sheep. In most circumstances, infection occurs when animals are in close physical contact and are more than 12 months old. Infection is established readily by subcutaneous injection, intradermal injection, and intratracheal application, but it does not occur after oral administration, with the exception of neonatal calves. Experimental transmission of the infection using tumor material, infected blood, or tissue culture virus can be achieved in cattle, sheep, and goats, and apparently also in chimpanzees, but the tumors are produced only in the three ruminant species. A sheep bioassay can be used to determine the presence of the virus in infected cattle. Risk Factors Animal Risk Factors The prevalence of infection based on seroprevalence is positively associated with increasing age in both dairy and beef cattle. The prevalence of infection in dairy cattle under 17 to 24 months of age is much lower than in adult cattle and increases sharply after 24 months of age when heifers join the milking herd and are in close contact with older cattle.11 The rate of spread may also be associated with the prevalence of infection; in herds with a prevalence of 13% to 22% when first tested, the spread was slow; in a herd with a prevalence of 42%, the spread was much more rapid. Genetic Resistance and Susceptibility.  Infection with BLV is not synonymous with clinical disease. Most animals that become infected do not develop neoplastic disease. Once infection has occurred, the subsequent development of only an antibody response, or antibody plus persistent lymphocytosis (PL), or antibody plus lymphosarcoma, with or without PL, is determined by the host’s genetic makeup. Lymphosarcoma, the clinically apparent form of BLV infection involving the clonal transformation of infected B cells, occurs in about 1% to 5% of BLV-infected cattle and seems to be under genetic control of the host.

787

Immune responsiveness and heritable resistance or susceptibility to infection are influenced by the host major histocompatibility complex (MHC).10 In cattle this MHC refers to the so called bovine lymphocyte antigen (BoLA). The earliest studies linked the presence of two class I BoLA-A alleles to the resistance to PL, an association that could not, however, be confirmed at the population level.10 Later studies found that resistance or susceptibility to PL was more closely associated with polymorphisms of the class II DRB3 gene. BoLA-A-DRB3 genes not only were correlated with resistance or susceptibility to PL and lymphosarcoma in Holstein–Friesian cattle, but also with the proviral load harbored by infected lymphocytes.10 A complex relationship exists among genetic merit, milk production, BoLA genotype, and susceptibility to PL. Cows with high genetic potentials for milk and fat yields are more susceptible to PL than cows with lower genetic potentials, but cows with PL do not produce yields of milk or fat according to their predicted genetic values. The major histocompatibility gene BoLA-A was found to be not only associated with resistance to persistent lymphocytosis, but also with the individual’s production potential. It was therefore hypothesized that genetic selection for increased milk production may have increased the susceptibility to BLV infection of dairy cows over the past decades.11 Early attempts to quantify the economic impact of subclinical infection emphasized differences in milk production between seropositive and seronegative cattle. This approach, however, is likely to be confounded by age differences and ensuing differences in the stage of the disease complex. Antibodies to BLV may be present in recently infected cows with no other abnormality, in cows over 3 years of age with PL, and in animals older than 6 years of age with tumors. Susceptibility to Other Diseases.  A highly significant correlation was shown between BLV infection and the persistence of Trichophyton verrucosum infection, which suggests that the immune system may be impaired in BLV-infected cows. Observations in Sweden indicate many significant associations between BLV infection status and measures of incidence, reproduction, and production, but most were of low magnitude. The risk for other infectious diseases seemed to be greater among BLV-infected herds, whereas the risk for noninfectious diseases did not differ. Immune Mechanisms Both humoral- and cell-mediated immunity are induced in natural BLV infection. Although BLV is primarily associated to B lymphocytes, BLV provirus has been detected in the DNA of CD2+ T cells, CD3+ T cells, CD4+ T cells, CD8+ T cells, monocytes, and granulocytes of infected but clinically healthy

788

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

animals.12 BLV infection was reported to disrupt the equilibrium between B and T lymphocytes not only in PL but also in AL BLV-infected cattle.12 Studies of cytokine profiles in healthy and BLV-infected cows suggest that type I and II cytokines are altered with increases in IL-10 and IL-4 concentrations and declines in IL-2, IL-12, and interferon-γ concentrations.13 Following infection, a persistent antibody response occurs primarily to the envelope glycoprotein gp51 and the major core protein p24 of the BLV virions. The time from infection to development of antibodies can be as long as 14 weeks. Experimental infection of calves with the virus results in seroconversion, which can be demonstrated with the ELISA in 4 to 5 weeks after infection. Lymphocytosis can occur at about the same time after infection. Environmental and Management Risk Factors Lack of Biosecurity The introduction of infected animals into a herd has a significant positive effect on the subsequent prevalence of infection and clinical disease. The appearance of new outbreaks of leukosis is almost always the consequence of the introduction of BLV-infected animals in farms or areas previously free of the infection. Calf Management The level of calf management in dairy herds is also a major risk factor. Any environmental factor or management practice that allows newborn calves access to infective blood will increase the level of infection in the calves, including prolonged close contact between the cow and calf immediately after parturition or the use procedures or instruments such as the following: • Gouge dehorners and ear-tagging equipment • Tattooing equipment • Instruments used for castration or the removal of supernumerary teats • Use of single needles for vaccination • Instruments for control of excessive fly population in calf barns Feeding colostrum of infected dams to newborn calves has been incriminated as potential route of infection in the past. More recent research indicates that administration of maternal colostrum to calves born from BLV-positive cows greatly decreased the risk of infection in early life compared with calves born to BLV-positive dams but not receiving colostrum from their mothers.8 These results suggest that exposure to BLV occurs around birth and is not dependent on ingestion of colostrum but that ingestion of colostrum-containing BLV antibodies lessens the risk of contracting BLV infection.8 Some observations have found positive associations between BLV status of dairy

herds and weaning age, housing preweaned calves in hutches or separate calf housing, and contact between young stock and older animals during the winter housing period. Pathogen Risk Factors BLV is highly cell associated and persists in a subpopulation of peripheral B lymphocytes and to a much lesser extent in subpopulations of T lymphocytes. Free virus is rarely or never found in the blood of infected cattle. EBL is therefore not highly contagious. Once an animal is infected, the virus DNA persists for life, incorporated into the DNA of infected lymphocytes. Economic Importance Economic losses resulting from BLV infection are associated with morbidity and mortality as a result of malignant lymphosarcoma, decreased productivity and longevity in clinically and subclinically infected cattle, BLV eradication or control measures, and restrictions of international trade. Losses associated with clinical disease can be economically significant at a herd level in high-prevalence herds but are generally not significant in herds with low BLV seroprevalence because only 0.1% to 5% of seropositive cows and 10% to 50% of cows with persistent lymphocytosis develop lymphosarcoma. Economic losses per case of lymphosarcoma in the United States were estimated to be $412.00 in a 2003 survey. In addition, malignant lymphosarcoma was found to be the largest single reason for carcass condemnation during postmortem inspection at slaughter plants in the United States, accounting for over 21% of all condemnations.14 The nature and extent of the economic losses associated with subclinical BLV infection have been controversial because the evidence has been conflicting and because the costs incurred by subclinical infection are difficult to assess. The effects of subclinical BLV infection on milk production, reproductive performance, longevity, and culling rate are variable. In some observations, a BLV-seropositive cow had a shorter life span than both its seronegative counterpart and the entire milk cow population.11,15 Among older dairy cows, BLV-seropositive cows were culled prematurely compared with uninfected cows. The culling rate was higher and milk production was lower in BLVinfected herds compared with BLV-free herds. The effect on reproduction was minor. In other observations, milk production, somatic cell count, age at disposal, and culling rate were not influenced by seropositivity.16,17 In a spreadsheet analysis of dairy herds in the Maritimes in Canada, total annual costs for an average infected 50-cow herd were $806.00 for EBL, compared with $2472.00 for Johne’s disease, $2412.00 for BVD, and $2304.00 for neosporosis.

The association between EBL infection and annual value of production on dairy herds in the United States, as part of the National Animal Health Monitoring System’s 1996 dairy herd study, found that compared with herds with no test-positive cows, herds with test-positive cows produced 218 kg less milk per cow and year, which is equivalent to approximately 3% of milk production. The mean annual value of production decreased by $1.28 for each 1% increase in herd seroprevalence (based on a milk price of $0.29/kg). When the effects of infection were examined according to genetic potential for milk and fat production in dairy cows, the results were surprising. BLV-infected cows with high genetic potential for milk and fat yields were found to be more susceptible to become affected by PL compared with cows with inferior genetic potential. At the individual cow level, infected cows had greater milk production than uninfected cows based on seropositivity to BLV and 305-day mature equivalent fat-corrected milk production. Among seropositive cows, those with PL were culled at a younger age and had reduced production in the last lactation relative to other groups. The cost of clinical disease and subclinical infection varied substantially with the prevalence of infection, whereas the cost of control varied with herd size. A basic BLV control program is considered economical in herds in which the prevalence of infection is greater than or equal to 12.5%. Trade Restrictions A major economic effect of the disease lies in import restrictions placed by countries free of EBL on infected cattle and on semen either from infected bulls or from noninfected bulls from a positive herd. It is the practice, particularly in countries that are recognized as free of EBL by the World Organization of Animal Health (OIE), to require proof of freedom from infection with the virus from animals about to be imported into the country. This is a matter of major importance when the cattle are purebred and are sold at high prices as breeding animals. Some countries are already demanding a negative blood test for all cattle and meat to be imported, and this could represent a loss of export markets for some countries. Zoonotic Implications The possibility of transmission of the virus from cattle to humans is a real one; the virus is commonly present in the milk of infected cows, and the disease has been transmitted to chimpanzees in this way. Using an immunoblot test, a serologic survey of 257 humans in California found at least one antibody isotype reactive with BLV in 74% of the sera tested. However, this does not necessarily mean that humans are actually infected with BLV. The antibodies could be a response to

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

heat-denatured BLV antigens from consumed milk or meat. Only 9% of the subjects indicated any direct contact with cattle or their biological products. There is an ongoing scientific debate on the possible association of leukemia in humans with exposure to BLV, either through direct contact with animals or carcasses or through consumption of dairy or meat products. A number of retrospective and prospective epidemiologic studies have explored the risk of leukemia among specific occupational groups, such as farmers and people working in food production or meat processing. Several of these studies reported a significantly increased risk of developing leukemia for workers in livestock farming or meat processing, whereas other failed to identify such an increased risk.18–21 Increased risk of developing leukemia in occupational groups having contact with cattle does not create an automatic association with exposure to BLV because people in these groups would share exposure to other chemical, biological, or environmental agents possibly causing or contributing to this risk. To this date in vivo studies have not provided any evidence that BLV increases the risk of disease in humans.19 Other Species Lymphosarcoma occurs sporadically in all species, but natural infection with the BLV virus has been demonstrated only in sheep and capybaras. Although there is no evidence of a relationship between bovine viral leukosis and any disease of pigs, there is a record of enzootic leukosis in that species, which is inherited.

PATHOGENESIS Virus and Lesion Infection with BLV virus may occur in utero, at the time of birth, or at a later stage of life and requires exposure of a susceptible

individual to infected lymphocytes. The virus primarily establishes a persistent infection in a subpopulation of B lymphocytes by integrating proviral DNA into the host cellular DNA. Other cells that were found to carry proviral DNA, although to a much lower extent, are CD2+ T cells, CD3+ T cells, CD4+ T cells, CD8+ T cells, γ/β T cells, monocytes, and granulocytes.11 The four possible outcomes after exposure of cattle to BLV are outlined in Fig. 11-7 and are as follows: 1. Failure of the animal to become infected, probably because of genetic resistance 2. Establishment of a permanent infection and the development of detectable levels of antibodies without clinical or hematological abnormalities (aleukemic infection) 3. Establishment of a permanent infection; the animal becomes seropositive and also develops PL, a benign lymphoproliferative process. It is not a preclinical stage of lymphosarcoma. 4. Infected, seropositive animals that may or may not have been through a stage of persistent lymphocytosis and that develop neoplastic malignant tumors—lymphosarcoma After infection, seroconversion occurs within 2 to 12 weeks, a time frame that, among other factors, is determined by the infective viral dose. Antibodies against BLV are not protective against tumor development. Whether or not the animal becomes infected or develops any of the other forms of the disease depends on the recipient’s genetic constitution. The outcome may also be influenced by the animal’s immune status and the size of the infective dose of virus. A subset of animals develops a persistent increase of lymphocytes that can become apparent any time after the infection but rarely occurs before 2 years of age. Persistent

lymphocytosis in affected animals persists for several years if not for life and may or may not precede the development of malignant lymphosarcoma. Persistent lymphocytosis in contrast to the malignant lymphosarcoma is the result of polyclonal proliferation of B lymphocytes and therefore presents a benign lymphoproliferative process. The increase of lymphocytes also involves an increase of the T lymphocyte count and a concomitant increase of the BLV antibody titer. Development of lymphosarcoma, a neoplasm of the lymphoreticular system, occurs in less than 5% of infected cattle and is the only clinically apparent form of EBL. This neoplasm is usually derived from a single cell clone and is never benign. Lymphosarcoma usually occurs in BLV-infected cattle 5 to 8 years of age. Lesions develop at varying rates in different animals so that the course may be quite short or protracted over several months. The outcome is invariably fatal. Lesions and Clinical Disease In adult cattle, almost any organ may be the site of lesions, but the abomasum, heart, and visceral and peripheral lymph nodes are most commonly affected. Depending on the organ that is most involved, several clinical syndromes occur. Involvement of the abomasal wall results in impaired digestion and persistent diarrhea. When the atrial wall is affected, congestive heart failure occurs. In nervous tissue, the primary lesion is in the roots of peripheral nerves and spreads along the nerve to involve meninges and cord. Involvement of the spinal meninges and nerves results in the gradual onset of posterior paralysis. The skin, reproductive tract, and periorbital tissues are commonly affected. In the cutaneous form, intradermal thickenings develop, which persist but do not cause discontinuity of the epithelium. They are composed of aggregations of neoplastic lymphocytes. Invasion of periorbital tissues commonly results in exophthalmos. Esophageal obstruction may result from mediastinal lymph node involvement in calves. The tumors consist of aggregations of neoplastic lymphocytes, but in many cases they may be more accurately described as reticulosarcoma. They are highly malignant and metastasize widely. The hemogram is variable, and although there may be an accompanying lymphocytosis, the presence of large numbers of immature lymphocytes in the blood smear is a more reliable indication of the presence of the disease. Some degree of anemia is common.

CLINICAL FINDINGS

Fig. 11-7  Possible pathways after exposure to bovine leukosis virus (percentage figures indicate proportion of seroconverted animals that develop the particular form referred to2).

789

This disease is characterized by tumors developing rapidly in many sites with an accompanying great variation in clinical signs and syndromes. An approximate indication of the frequency with which individual signs appear is set out in Figure 11-8.

790

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

variable appetite, persistent diarrhea, not unlike that of Johne’s disease, and occasionally melena as a result of bleeding of an abomasal ulcer. Tumors of the mediastinal nodes may cause chronic, moderate bloating.

Fig. 11-8  Clinical diagnosis frequency of predominant signs of bovine leukemia—1100 field cases. (Courtesy of Canadian Veterinary Journal).

The usual incubation period is 4 to 5 years. This form is rarely seen in animals under 2 years of age and is most common in the 4- to 8-year-old age group. Persistent lymphocytosis without clinical signs may occur earlier but rarely before 2 years of age. Many cows remain in the preclinical stage for years, often for their complete productive lifetime, without any apparent reduction in performance, but clinical disease appears in a small subset of these cows. The clinical signs and the duration of the illness vary with the number and importance of the sites involved and the speed with which the tumor masses grow. In 5% to 10% of clinical cases the course is peracute, and the affected animals often die suddenly or unexpectedly without any prior evidence of illness. Involvement of the adrenal glands and rupture of an abomasal ulcer or an affected spleen, followed by acute internal hemorrhage, are known causes. These animals are often in good bodily condition. In most cases the course is subacute (up to 7 days) to chronic (several months) and initiated by an unexplainable loss of body condition and appetite, pallor, and muscular weakness. The heart rate is not increased unless the myocardium is involved, and the temperature is normal unless tumor growth is rapid and extensive, when it rises to 39.5° to 40° C (103° to 104° F). Although the following specific forms of the disease are described separately, any combination of them may occur in any one animal. In many cases, clinical illness sufficient to warrant the attention of the veterinarian is not observed until extensive involvement has occurred and the possibility of slaughter of the animal for beef purposes cannot be considered. On the other hand, many cases are examined at a time when diagnostic clinical signs are not

yet evident. Once signs of clinical illness and tumor development are detectable the course is rapid, and death occurs in 2 to 3 weeks. Enlargement of the Superficial Lymph Nodes Enlargement of the superficial lymph nodes occurs in 75% to 90% of clinical cases and is often an early clinical finding. This is usually accompanied by small (1 cm in diameter) SC lesions, often in the flanks and on the perineum. The skin lesions are probably enlarged hemolymph nodes and are of no diagnostic significance, often occurring in the absence of other signs of the disease. In many cases with advanced visceral involvement, peripheral lesions may be completely absent. Enlargement of visceral lymph nodes is common, but these are usually subclinical unless they compress other organs such as intestine or nerves. They may be palpable on rectal examination, and special attention should be given to the deep inguinal and iliac nodes. In advanced cases, extensive spread to the peritoneum and pelvic viscera occurs, and the tumor masses are easily palpable. Although the enlargement of lymph nodes is often generalized, in many cows only a proportion of their nodes are involved. The enlargements may be confined to the pelvic nodes or to one or more SC nodes. Involvement of the nodes of the head is sometimes observed. The affected nodes are smooth and resilient and in dairy cows are easily seen, and their presence may be marked by local edema. Occasionally, the entire body surface is covered with tumor masses 5 to 11 cm in diameter in the SC tissue. Digestive Tract Lesions Digestive tract lesions are common. Involvement of the abomasal wall results in a

Cardiac Lesions Lesions in the heart usually invade the right atrial wall primarily, causing right-side congestive heart failure. There is hydropericardium with muffling of the heart sounds, hydrothorax with resulting dyspnea, engorgement of the jugular veins, and edema of the brisket and sometimes of the intermandibular space. Tachycardia as a result of insufficiency and arrhythmia as a result of heart block are common. A systolic murmur is also common, along with an abnormal jugular pulse. The liver may be enlarged and palpable caudal to right costal arch, and passive congestion of the liver and visceral edema results in persistent diarrhea. Nervous System Involvement Neural lymphomatosis is usually manifested by the gradual onset over several weeks of posterior paralysis. Knuckling of the fetlocks of the hindlegs while walking is common, and one leg may be more affected than the other. This is followed by difficulty in rising, and finally by clinical recumbency and inability to stand. At this stage, sensation is retained, but movement is limited or absent. There may be a zone of hyperesthesia at the site of the lesion, which is usually at the last lumbar or first sacral vertebra. Appetite and other functions, apart from the effects of recumbency, are usually normal. Metastases in the cranial meninges produce signs of space-occupying lesions with localizing signs referable to the site of the lesion. Less Common Lesions Less common signs include enlargement of the retropharyngeal lymph nodes, which may cause stertor and dyspnea. Sometimes clinically detectable lesions occur in the periorbital tissues, causing protrusion of the eyeball (exophthalmos), and in the limb muscles, ureter and kidney, and genitalia. Involvement of the uterus may be detectable as multiple nodular enlargements on rectal examination. Severe bilateral exophthalmos may occur, along with generalized lymphadenopathy. Periureteral lesions may lead to hydronephrosis with diffuse enlargement of the kidneys, whereas tumors in renal tissue cause nodular enlargements. In either case terminal uremia develops. BLV particles have been detected by electron microscopy around lymphocytes in the mammary tissue of BLV-antibody-positive cows affected by subclinical mastitis. Whether the virus is a causative agent or an immunosuppressant in bovine mastitis is unknown.

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

Other Species Outbreaks of lymphosarcoma in sheep have been observed with clinical, epidemiologic, hematological, and necropsy findings similar to those of enzootic bovine leukosis. B-cell leukemia has been described in sheep. Infection of other species with BLV has not been demonstrated, but epidemic occurrences of lymphosarcoma have been observed in pigs, but only sporadic cases in horses.

CLINICAL PATHOLOGY

A definitive antemortem diagnosis depends on the clinicopathological examination of the animal. Several diagnostic techniques are available, and it is important to make the appropriate selection for the particular stage of the disease that is being considered, as follows: • Diagnosis of the viral infection is made by serologic or virological techniques. • Persistent lymphocytosis is identified by hematology but is not pathognomonic for BLV infection. • Neoplastic tumors are identified by histologic examination of a biopsy specimen. Because of the increasing economic impact of BLV infection in the cattle industry, the availability of a highly sensitive and specific assay for the identification of BLVinfected animals is of critical importance. Such an assay is needed for the selection of BLV-free cattle for commercial sale, prepurchase testing of breeding animals and import or export testing, and control and eradication programs. Ideally, the assay should be practical, inexpensive, and able to be adapted for large-scale use. Diagnosis of the Presence of Infection With BLV Serologic Tests Seroconversion occurs between 3 and 16 weeks postinfection. Virtually all cattle infected with BLV will continuously have antibodies against the major internal (p24) and envelope (gp51) virion proteins in their serum, and serologic tests are commonly used for the diagnosis of BLV infection in cattle over 6 months of age. Maternally derived antibodies may be detectable until 7 months of age and are indistinguishable from antibodies resulting from infection. BLV antibodies tend to decline in the periparturient period as a result of the shift from the dam’s circulation to the mammary gland, and titers may drop below the detection limit from 2 to 6 weeks before to 2 weeks after calving.22 A number of diagnostic tests are available to diagnose seroconversion in individual animals and on a herd level, as discussed in the following sections. Enzyme-Linked Immunosorbent Assay in Serum or Milk.  In the last decade ELISAbased testing has replaced the AGID in

eradication programs in several countries and is one of the prescribed diagnostic tests for international trade. It is more sensitive than other serologic tests and can be used on milk. The ELISA can detect antibody titers 10- to 100-fold below the detection limit of the AGID. The superior sensitivity of the ELISA allows detection of antibodies in pooled serum samples of herds with a prevalence of less than 1%, whereas the AGID test detected only 50% of the herds detected by the ELISA. Two commercially available ELISAs and the PCR were evaluated and compared with the AGID to detect antibodies to BLV or its nucleic acid. The ELISA tests detected about 10% more reactors than the AGID and the electrophoretic immunoblotting results. Some ELISA-positive animals were not detected by the PCR. Four commercially available BLV-ELISA kits from Europe and the United States were compared with the AGID test officially approved by the Canadian Food Inspection Agency. The ELISA tests were more sensitive than the AGID test kits. A highly sensitive and specific blocking ELISA comparable to the radioimmunoprecipitation assay for the detection of BLV antibodies in serum and milk samples has been developed. The milk ELISA has been adopted for testing milk from individual cows and pooled milk samples. A comparison of the ELISA and AGID tests for the detection of BLV antibodies in bovine serum and milk found a high level of agreement. The bulktank milk ELISA is useful for identification of herds that are negative for BLV infection and to monitor BLV-negative herds. The antibody level in milk is lower than in serum but the sensitivity of the ELISA is as effective as for sera. Testing of bulk milk is a useful and practical method for large-scale epidemiologic studies and initial eradication programs. Heifers, bulls, and dry cows and youngstock over 1 year of age that are not included when bulk milk is tested need to be sampled individually before a herd is declared free of the virus. The sensitivity and specificity of the milk ELISA is estimated to be adequate until the prevalence of BLVinfected individuals in the country is less than 1%. Herds identified as positive by the milk ELISA would require further testing at the individual or herd level to definitively establish their BLV status. Agar Gel Immunodiffusion Test.  The AGID is a specific but not very sensitive diagnostic test for BLV. It has, however, been proven to be highly useful and efficient as a basis for eradication or control programs because it is simple, easy to perform, and inexpensive.22 It remains one of the prescribed tests for international trade. Most commercial AGIDs test for the presence for both antibodies against p24 and gp51 but are not standardized for the demonstration of antibodies against gp51.

791

Radioimmunoprecipitation Assay.  The radioimmunoprecipitation (RIP) assay, which uses gp51 or p24 as antigen, is a highly sensitive and specific method for serologic diagnosis of BLV infection. The RIP assay has been used as the criterion-referenced standard to critically evaluate the performance of other diagnostic tests for BLV infection. Detailed comparisons of various BLV assays in a large number of cattle of various origins and ages found that the RIP assay is the most sensitive and specific test. However, its major disadvantage is that it requires a gamma counter and radioisotopes, which are expensive. Radioimmunoassay.  Radioimmunoassay (RIA) is suitable for individual cow testing because of its accuracy. There are several versions of this test, and the one using the virion gp antigen is preferred. It is one of the most sensitive tests and is useful for the detection of BLV antibodies in cattle exposed no longer than 2 weeks, in milk samples, and in serum samples from periparturient dams. Detection of Virus Polymerase Chain Reaction.  The PCR is a sensitive and specific assay for direct diagnosis of BLV infection in peripheral blood lymphocytes. The test is useful for the early detection of BLV infection even before antibodies are present. It is more sensitive than the ELISA or AGID in detecting infected cattle where the prevalence of infection is less than 5%. The test can identify proviral DNA of BLV in the lymphocytes of calves at birth in calves born to infected cows. At birth, the presence of an antibody titer can result from passive transfer of immunity or perinatal infection, and the PCR test can differentiate uninfected newborn calves with colostral antibodies from BLV-infected calves and detect the presence of the virus in the presence of antibodies. The PCR has a practical application in the identification of BLV-infected calves, regardless of colostral antibody, which allows immediate removal of the source from the herd. In a dairy herd with a high prevalence of BLV, a positive PCR assay result provided definitive evidence that a cow was infected with BLV. However, sensitivity and specificity were 0.672 and 1.00, respectively. The predictive value of a positive test was 1.00, and the predictive value of a negative test was 0.421. Thus PCR assay alone is unreliable for routine detection of BLV in herds with a high prevalence of BLV infection. The PCR can also be used to ensure that cattle used in the production of a wholeblood vaccine for tick-borne disease are free from BLV infection. The sheep bioassay, currently in use, requires 4 months of serologic testing to ensure that donor animals are not infected. Replacement of the sheep bioassay with the PCR could result in considerable saving of time and effort. Use of the PCR

792

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

requires stringent precautions to prevent false-positive results from contamination of samples with PCR product. A nested PCR identified 98% of BLVseropositive cows from blood and 65% from milk, whereas real-time PCR detected 94% of BLV-seropositive cows from blood and 59% from milk. BLV was also detected in 10% of seronegative cows, most likely because of early detection before seroconversion. Differentiation Between Enzootic and Sporadic Bovine Leukosis.  The role of BLV in some cases of sporadic bovine lymphomas (SBL) needs to be reexamined. The findings of persistently seronegative PCRpositive and seropositive PCR-negative cattle indicates that the BLV cannot be excluded as a causative agent in some cases of SBL. Enzootic bovine leukosis cannot be distinguished from SBL on histopathological examination. The ELISA is recommended as a method of choice to differentiate between EBL and SBL because it is a rapid, reliable, and sensitive test that is inexpensive and easy to perform. In cases where no blood or other fluids are obtained, the PCR test is the most useful method for the direct detection of BLV. Diagnosis of Persistent Lymphocytosis Approximately 30% of animals infected with BLV develop PL, which is defined as a benign increase in the absolute lymphocyte count of three or more standard deviations above the normal mean as determined for that respective breed and age group of animals in leukosis-free herds. The PL is an increase in peripheral B lymphocytes. It has been suggested that one additional criterion for PL should be that it persists for more than 3 months. When PL was first recognized in herds that experienced malignant lymphosarcoma, it was considered a subclinical expression of the tumor stage of the disease. Although persistent lymphocytosis is not pathognomonic for BLV infection, it became an important diagnostic criterion in control and eradication programs until BLV was identified as causative agent and serologic tests became available to more accurately identify infected animals. The majority of cells involved in PL are normal lymphocytes, but atypical and abnormal forms have been described and are considered as indicative of preleukemic condition. The total count increases from a normal of 6000 to as high as 15,000/µL. An increase in the percentage of lymphocytes in the total white blood cell count from the normal of 50% to 65% is considered a positive result. The presence of 25% or more of the total lymphocyte count as atypical immature cells is also considered a significant aberration. The PL may or may not subside in animals that subsequently develop lymphosarcoma.

The association between the strength of serologic recognition of BLV by the use of ELISA and lymphocyte count in BLVinfected cows has been examined. The sample-to-positive ratio, which is the ratio between the test sample and a positive control sample, was compared among lymphocytotic and nonlymphocytotic cows. The sample-to-positive ratio and lymphocyte count were related, but cows with high sample-to-positive ratio were not always lymphocytotic. Culling cows on the basis of sample-to-positive ratio will reduce culling of ELISA-positive cows; however, culling on the basis of lymphocyte count will eliminate a greater proportion of the reservoir of infection. Diagnosis of Lymphosarcoma Lymphosarcoma can only be diagnosed by histopathological examination of a section of tumor material obtained by biopsy or necropsy. A needle aspirate of an enlarged peripheral lymph node may provide a rapid and inexpensive diagnosis. Enlarged lymph nodes or hemolymph nodes are the usual sources, but when the genital tract is involved an exploratory laparotomy is usually performed so that a sample can be obtained. The lymphocyte count may increase to 20,000 to 30,000/µL and in some cases may reach values of 50,000 to 100,000/µL, and even 400,000 to 500,000/µL. Conversely, in some cases, the lymphocyte count decreases. Chromosomal changes may be detectable in cells from lymph nodes or in leukocytes from peripheral blood of affected animals. When there is myocardial involvement there may be obvious changes in the electrocardiogram, but these are unlikely to be of value in differential diagnosis.

NECROPSY FINDINGS

Enzootic bovine leukosis is mostly a disease of adult cattle characterized at necropsy by markedly enlarged lymph nodes and multiple firm, white tumor masses in any organ but especially the liver, spleen, heart, abomasum, and spinal cord. Affected lymph nodes may be enormously enlarged and be composed of both normal and neoplastic tissue. The latter is firmer and whiter than normal lymphoid tissue and often surrounds foci of bright yellow necrosis. Enlarged lymph nodes may appear anywhere in the body but are common in the retrobulbar, pharyngeal, and pelvic areas. An affected liver may have discrete nodular masses, or it may be diffusely enlarged and pale, and can be easily misinterpreted as fatty degeneration rather than a neoplastic process. In the heart, the tumor masses invade particularly the right atrium, although they may occur generally throughout the myocardium and extend to the pericardium. The frequency of early changes in the subepicardial tissue of the right atrium suggests that this is an area from which tissues should be selected in latent or

doubtful cases. The abomasal wall, when involved, shows a gross, uneven thickening, with tumor material in the submucosa, particularly in the pyloric region. Similar lesions occur commonly in the intestinal wall. Deep ulcerations in the affected area are not uncommon. Involvement of the nervous system usually includes thickening of the peripheral nerves coming from the last lumbar or first sacral cord segment or more rarely in a cranial cervical site. This may be associated with one or more circumscribed thickenings in the spinal meninges. Less common sites include the kidney, ureters, and uterus. Histologically, the tumor masses are composed of densely packed, monomorphic lymphocytic cells. The cleaved variant of the diffuse large cell type with a high mitotic index is characteristic of enzootic lymphoma, and this high-grade type of B-cell tumor may be a consequence of the viral etiology of this form of the disease. It is possible to confirm viral infection in some cases by a PCR test, but such testing is rarely justified. Samples for Confirmation of Diagnosis • Histology—formalin-fixed samples of gross lesions, plus enlarged lymph nodes, bone marrow, liver, spleen, thymus, right atrium, abomasum, uterus (LM, IHC) • Virology—neoplastic tissue (PCR) DIFFERENTIAL DIAGNOSIS Because of the very wide range of clinical findings, a definitive diagnosis of bovine leukemia virus (BLV) is often difficult. Enlargement of peripheral lymph nodes without fever or lymphangitis is unusual in other diseases, with the exception of tuberculosis, which can be differentiated by the tuberculin test. In the absence of these enlargements, the digestive form may easily be confused with Johne’s disease. The cardiac form closely resembles traumatic pericarditis and endocarditis, but there is absence of fever and toxemia, and the characteristic neutrophilia of these two diseases is usually absent. Involvement of the spinal nerves of meninges may be confused with spinal cord abscess or with the dumb form of rabies. An examination of cerebrospinal fluid may be of value in determining the presence of an abscess, and rabies has a much shorter course and other diagnostic signs. Multiple lymph node enlargements in the abdominal cavity and nodular lesions in the uterine wall may be confused with fat necrosis, but the nature of the lesion can usually be determined by careful rectal palpation. Stertor caused by enlargement of the retropharyngeal lymph nodes is also commonly caused by tuberculosis and actinobacillosis. Cases of sporadic bovine leukosis that are BLV-negative may resemble lymphosarcoma of enzootic bovine leucosis.

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

TREATMENT

VetBooks.ir

There is no treatment.

CONTROL

The disease can be eradicated from a herd, and even a country, or controlled at a low level. The option chosen depends initially on the prevalence of infection in the herd, the value of the animals in the herd, and whether a governmental indemnity offered for seropositive cows that are culled and sent to slaughter. History of Compulsory Eradication Programs in Europe Control and eradication programs have been in effect on a nationwide basis in several western European countries. Denmark began an eradication program in 1959 based on the occurrence of clinical lymphosarcoma and the identification of cattle with PL using the Bendixen hematological key for classifying cattle as normal, suspect, or lymphocytic. This eradication program was established more than 10 years before identification of the etiologic agent of the disease. Even though the causative agent was unknown, bovine lymphosarcoma was widely accepted to be an infectious disease because of its occurrence in clusters of diseased cows in certain herds.1 Leukosis was declared a reportable disease, and all adult cattle from herds in which cases of leukosis originated were subjected to a hematological examination. Affected herds were quarantined, and indemnity was offered to induce owners to have their entire herd slaughtered. This herd-slaughter policy was continued until 1982. When the AGID test became available, the Bendixen key was discontinued, and only the AGID test was used in the official program between 1979 and 1982. Routine testing was discontinued in 1982. Surveillance involved testing random sera collected from every sixth adult cow to be slaughtered. According to the official Danish control program, the incidence of tumors in adult cattle at the start of the eradication program was at least 10 times greater than that 10 years later. The hematological test was less sensitive than subsequent serologic tests but the specificity was fairly high, and only a few herds were erroneously classified as leukosis herds. When the serologic tests were introduced, some herds that were classified as leukosis-free based on the hematological Bendixen key were found to be infected. In Britain a national testing program was begun in 1992 that led to successful eradication of the disease. All blood samples collected for routine periodic testing for brucellosis have also been tested for BLV, and milk samples are collected every 3 months from dairy herds for BLV testing. The prevalence of infection has been low and the source of infection undetermined. Some of the animals had been imported from Canada, but in other cases there was no association with importation.

Enzootic bovine leukosis was eradicated from Finland in 1996. The disease was first recognized in 1966, and it required 30 years of use of the key principles of test and slaughter policy to achieve eradication. The infection status was monitored at meat inspection, hematologically between 1970 and 1977, and serologically between 1978 and 1989. Annual surveys including all dairy herds and samples from beef animals were conducted from 1990 to 2001. Bulk-tank samples represented the dairy herds in the surveys; beef animals were sampled individually at slaughter. The maximum positive herd-level percentage in the survey was 0.03%. The herd level prevalence of infection never exceeded 5%. Considering the animal-health aspects and possible consumer reactions against having a widespread retrovirus infection in food-producing animals, and the requirements for exporting cattle and semen, Sweden introduced a national program for the eradication of BLV in 1990. An ELISA test was evaluated for detection of antibodies to BLV in individual and bulk milk and serum samples. In the meantime, a total of 17 countries of the European Union are officially free of BLV. Similarly, since 2010 New Zealand, after having implemented a BLV eradication program, is free of evidence of EBL, and Australia expects to have eradicated the disease. In Canada and the United States, it is considered cost prohibitive to cull and slaughter all seropositive cattle because of the high prevalence of infection. Many seropositive cows are valuable pedigreed animals, and there are no indemnity programs available. Thus all control and eradication programs in these countries are herd based and strictly voluntary. Livestock producers are willing to adopt control programs because of the economic losses associated with export restrictions if their cattle are infected and the losses resulting from the occasional clustering of cases of lymphosarcoma. Eradication Programs Enzootic bovine leukosis can be eradicated only by the following methods: • Test and slaughter of cattle infected with the virus—programs based on the culling of seropositive cows are effective. • The maintenance of a closed herd, which permits the entry of only those animals free of infection The efficiency of such a program depends on the accuracy of the test used to identify the infected animals and the repetition of the test at an appropriate interval so that animals that were in the incubation stages of the disease at the time of the first test will have had time to seroconvert. The recommended procedure is as follows: 1. Identify infected animals using the serum or milk ELISA or AGID test. 2. Cull and slaughter seropositive animals immediately.

793

3. Retest the herd 30 to 60 days later. 4. Use the PCR assay to test young calves and as a complementary test for clarifying doubtful test results in herds with a low prevalence of infection. Testing is repeated until the herd has a negative test. When the herd is negative, testing is repeated every 6 months and the herd declared free when there have been no positive reactors for 2 years. Future introductions into the herd are managed most safely by artificial insemination or fertilized ovum transfer, or importations of animals that have been tested and are seronegative on two tests carried out 30 and 60 days before arrival. In herds where the prevalence is high, a two-herd scheme can be successful. Newborn calves are removed from seropositive cows immediately after birth, fed colostrum from seronegative cows, and raised in isolation. All animals over 6 months of age are tested periodically and seropositive animals culled. The parent herd is eventually disposed of as negative replacement animals become available. Only those bulls that are seronegative may be used, and they must be tested every 3 months. Although eradication is biologically feasible, it is unlikely that area eradication programs will be implemented on an extensive scale because losses from the disease are not sufficiently high, and there is a high risk of insect vectors reintroducing it, which poses a real threat to maintenance of a BLV-free herd. The cost-effectiveness of an eradication program on a national basis would be a major consideration. For an individual herd, it is feasible, provided some steps are taken to increase the genetic resistance of the residual stock and to reduce the chances of incontact infection occurring. Limitation of Spread of Infection In herds with a high prevalence of infection, the test and slaughter method of eradication is not economically viable if the animals have a high economic value because of superior genetic potential. Control of infection in these herds is possible using embryo transfer from infected dams to negative recipients and isolation of newborn calves, but these are not practical on a country-wide basis. An alternative method is segregation of BLV-infected and noninfected animals based on the serum/milk ELISA or AGID test. This is known as the test and segregation method, which is based on the evidence that the spread of infection between animals is relatively slow and that the virus is spread by movement of infected animals from one herd to another and within a herd. Following the initial testing of the herd, the herd is divided into two groups, BLV-positive and BLV-negative animals, and kept at least 200  m apart. A third separate location is used for quarantine of replacement animals.

794

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Replacement animals must be found negative in two consecutive serum antibody tests, the first within 30 days before purchase and the second after 60 days of isolation, before being moved into the negative group. The serologic tests are conducted every 3 months, and the reactors removed to the positive group location until the remaining animals in the herd have attained BLV-negative status by the test. Thereafter, the tests are done every 6 months and continued until at least four consecutive negative tests are obtained for each herd. Variations of this method of test and segregation with subsequent removal of seropositive animals in the routine culling program have been successful. The colostrum and milk fed to calves in the BLV-negative group must be from seronegative cows or be pasteurized to inactivate the virus. In Canada, cattle owners may enroll in the Canada Health Accredited Herd program to declare freedom from EBL. All reactors must be removed from the herd. If a large number of reactors are detected, two herds on two separate farms can be established: one herd comprised of the reactors and the other of cattle that are seronegative. Calves from the reactor herd can be added to the accredited herd in accordance with strict isolation and testing procedures. To qualify for accredited certification, a herd must have two consecutive negative herd tests, at least 4 and less than 12 months apart. The tests must include all cattle in the herd. The first annual renewal test must occur no more than 12 months following the second qualifying test for certification, and must include all cattle in the herd. Subsequent renewal tests must occur within the same 12-month interval. Only cattle 24 months of age and older must be tested, but a herd inventory and audit must be performed on the whole herd. In herds with reactors, the two qualifying tests do not begin until at least 4 months after the removal of the last reactor uncovered during any test. Herd additions can be made during the qualifying test period or after certification has been achieved. Each animal must be accompanied by a health certificate, and depending on the enzootic bovine leukosis status of the originating herd, certain testing and isolation procedures could apply. Owners wishing to have their animals attend exhibitions can do so providing they adhere to certain conditions. Properly processed semen and embryos can be introduced without restrictions. Owners are encouraged to follow preventive health management practices to augment the eradication of enzootic bovine leukosis from their herds. These include all areas where blood transfer could occur (needles, dehorning, castrating, extra teat removal, ear-tagging, tattooing, hoof trimming, rectal palpations, drenching) and other procedures that transfer leukocytes, in addition to routine insect control.

Prevention of Infection in Calves and Young Stock Several management techniques can be used to prevent infection in calves from birth until they become herd replacements. Immediate removal of newborn calves from the maternity pen and feeding of colostrum and milk from seronegative cows is widely accepted as effective in preventing infection in calves. Postnatal infection in calves can also be minimized by feeding milk replacer and/or whole milk from noninfected cows. The use of colostrum and milk from noninfected cows permits early serologic detection of infected calves. However, feeding colostrum from seropositive cows to newborn calves can provide significant protection from infection during the first 3 months of life. Field studies indicate that colostrum-derived BLV antibodies may prevent as much as 50% of the infection during the first 3 months compared with calves that did not receive colostrum with BLV antibodies. Further reduction in the risk of infection via colostrum can be achieved by pasteurization of the colostrum at 63° C (147° F) for 30 min. The colostrum-derived BLV antibodies will, however, delay early detection of infection in calves. The replacement of whole-milk feeding with high-quality milk replacer may also be considered. Transmission to newborn calves can also be reduced by avoiding exposure to maternal blood at the time of parturition, housing calves in individual hutches with individual feeders and waterers, and management techniques to avoid iatrogenic transmission. When handling a group of calves, the youngest ones should be handled first and the older and sick calves last. Equipment that could act as a fomite in transferring blood should be disinfected with chlorhexidine when used between calves. These instruments include the following: • Nose tongs • Scissors • Forceps • Dehorning instruments • Esophageal tubes • Balling guns • Tattoo equipment • Ear taggers Dehorning of calves with the electrocautery method before 2 months of age can reduce the prevalence of infection compared with gouge dehorning, which allows the transfer of infected blood between calves. Handling facilities that become contaminated with blood should be cleaned between calves. Fly control should be instituted as necessary. Single needles should be used for vaccination, and calves should be tested serologically for BLV infection at about 6 months of age. A marked reduction in the prevalence of infection within the heifer age groups of a dairy herd with a high prevalence can be achieved by the following practices:

1. Use of single needles and individual sleeves for rectal examination 2. Disinfection of tattoo equipment before use 3. Dehorning by use of electrocautery Biosecurity Prevention of entry of infection into herd can be achieved by ensuring that all imports into the herd have been tested at least 30 days before arrival and are seronegative. Control of insect vectors is highly desirable. Blood transfusions and vaccines containing blood, such as those used for babesiosis and anaplasmosis, are particularly potent means of spreading the disease, and donors must be carefully screened to ensure that they are free of the disease. In the future, the selection of cattle with inherent resistance to BLV may be a possibility. Embryo transfer from valuable pedigreed seropositive cows may aid in reducing prenatal infection. Insemination is not a method of transmission, and thus artificial breeding programs are not disrupted. Vaccine The possibility of a vaccine for BLV has been explored extensively. Thus far inactivated virus vaccines, cell-derived vaccines, viral subunit vaccines, recombinant vaccinia virus vaccines, and synthetic peptides have been examined, without much success.10 REFERENCES

1. George JW. Vet Clin Pathol. 2007;36:220-221. 2. Commission of the European Union. 2012 At: ; Accessed 21.10.2013. 3. USDA-APHIS. 2009 At: ; Accessed 21.10.2013. 4. USDA-APHIS. 1997 At: ; Accessed 21.10.2013. 5. Scott HM, et al. Can Vet J. 2006;47:981-991. 6. Van Leeuwen JA, et al. Can Vet J. 2006;47:783-786. 7. Gutiérrez G, et al. Vet Microbiol. 2011;151:255-263. 8. Nagy DW, et al. J Vet Intern Med. 2007;21:1104-1107. 9. Erskine RJ, et al. J Dairy Sci. 2012;79:445-450. 10. Rodriguez SM, et al. Viruses. 2013;1210-1248. 11. Bartlett PC, et al. J Dairy Sci. 2013;96:1591-1597. 12. Panei CJ, et al. BMV Vet Res. 2013;9:95. 13. Erskine RJ, et al. Am J Vet Res. 2011;72:1059-1064. 14. USDA-FSIS. 2002 At: ; Accessed 21.10.2013. 15. Erskine RJ, et al. J Dairy Sci. 2012;95:727-734. 16. Tiwari A, et al. J Dairy Sci. 2007;90:656-659. 17. Sorge U, et al. J Dairy Sci. 2011;94:5062-5064. 18. Polychronakis I, et al. J Occup Med Tox. 2013;8:14. 19. Neasham D, et al. Occup Environ Med. 2011;68:77-81. 20. Cocco P, et al. Int J Cancer. 2012;132:2613-2618. 21. ‘t Mannetje A, et al. Occup Environ Med. 2008;65:354-363. 22. OIE Terrestrial Manual. 2012. Chapter 2.4.11, At: , (Accessed 21.10.2013).

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

EQUINE INFECTIOUS ANEMIA (SWAMP FEVER) SYNOPSIS Etiology Equine infectious anemia virus, a retrovirus (lentivirus). Epidemiology Worldwide distribution. Affects all species of Equidae. Transmission of disease is by contaminated blood of clinically affected or inapparently infected horses during interrupted feeding of blood-feeding insects or iatrogenically. Horses are infected for life. Clinical signs Fever, depression, edema, petechial hemorrhages, abortion, chronic weight loss, splenomegaly. Clinical pathology Anemia, thrombocytopenia, hypergammaglobulinemia, positive agar gel immunodiffusion (AGID) or competitive enzyme-linked immunoassay (cELISA)   test. Diagnostic confirmation AGID or cELISA test. Differential diagnosis list: Acute disease:

• • • • • • • •

Purpura hemorrhagica Babesiosis Equine granulocytic ehrlichiosis Equine viral arteritis Autoimmune hemolytic anemia Leptospirosis Parasitism Idiopathic thrombocytopenia

Chronic disease:

• Neoplasia • Internal abscessation • Liver disease

ETIOLOGY The equine infectious anemia virus (EIAV) is a retrovirus, a member of the subfamily Lentivirinae of the family Retroviridae. It is an RNA virus that uses a reverse transcriptase enzyme to generate proviral DNA, which is spliced into the host’s genome.1 The virus infects only Equidae, and there is no evidence that it infects or causes disease in humans. EIAV shares antigenic crossreactivity with the human and feline immunodeficiency viruses but not with the viruses causing caprine arthritis–encephalitis or maedi–visna of sheep. The EIAV genome is composed of three major genes: gag—encoding matrix and capsid proteins; pol—encoding proteases, reverse transcriptase, and integrase; and env—encoding envelope glycoproteins.2 Antibodies against gag-encoded proteins (e.g., p26) are used commonly for diagnosis of equine infectious anemia (EIA) by AGID or ELISA testing. Analysis of the genome of the gag gene has revealed a strong geographic localization of clades of EIAV that are clearly related to

the evolutionary history of modern horses. Both horses and EIAV appear to have a Eurasian origin that might have differentiated as a consequence of horse movement associated with human migration. Analysis of the gag genome reveals that EIAV strains from the Americas form a distinct group with a potentially single common origin temporally associated with European colonization of North America.3 The close geographic structuring of EIAV is a result of the insect-borne nature of the infection, which favors transmission over short distances as opposed to long distances, and the limited mobility, until very recently, of equids.3 There are three distinct monophylogenetic groups, two of which (EIAV-1 and EIAV-2) contain only European strains and the third of which (EIAV-3) contains European, Asian, and American strains. This third group has three clades including European, Asian, and all American strains. The two Japanese strains in this group are of American origin.4 Genomic characterization of EIAV enables a more complete understanding of the geographic origin of virus strains and the epidemiology and pathogenesis of the disease. For instance, analysis of strains of the virus isolated in Belgium in 2010 confirmed that the source is Romanian,5 that Japanese strains V70 and V26 are of not of Japanese origin, that there exists a distinct Japanese strain,4,6 and that the virus that caused the 2006 outbreak in Ireland was of European strain origin.7 There is considerable antigenic drift in the surface glycoproteins (gp45, gp90), and the emergence of novel antigenic strains within an individual horse is associated with the relapsing febrile reactions characteristic of the disease. Examination of variations in the viral regulatory protein, Rev, and the transmembrane protein, gp90, demonstrates the existence of viral quasispecies such that genetically distinct viral subpopulations of differing phenotype exist within a chronically infected, often asymptomatic, animal. Mutations in gp90 are driven by the host immune response and can cause relatively large insertions or deletions in the gene.8 EIAV, like all retroviruses, integrates into the genome of the host by insertion of viral cDNA into the host DNA. This process involves nonrandom insertion of viral cDNA, as a provirus, into the host genome in the form of preintegration complexes. This integration of viral DNA into the host genome is lifelong. The sites of EIAV insertion into the genome of infected horses have been described.1

EPIDEMIOLOGY Occurrence EIA has been diagnosed on all continents except Antarctica, and there is increasingly detailed information on distribution of the virus and infection rates in many countries

795

and localities, including Turkey,9–12 Brazil,13–16 Oman,17 Ethiopia,18 Pakistan,19 Costa Rica,20 Romania,21 Taiwan (where the disease is not currently present),21 Greece,22 India,22 Italy,23 and Mongolia,23 among others. In Europe it is most prevalent in the northern and central regions. It has appeared in most states of the United States and the provinces of Canada, but the principal enzootic areas are the Gulf Coast region and the northern wooded sections of Canada. Extensive serologic surveys using the AGID (Coggin’s) test have shown rates of infection of 1.5% to 2.5% in the United States, 6% in Canada, a low level in France, 1.6% in West Germany, and 15% to 25% in Argentina. Within a geographic area, the prevalence of infection (positive AGID) varies depending on the density of the population, the proportion of carrier animals, and the density of the population of insect vectors. Under ideal conditions the incidence of infection can approach 100% over a period of weeks, but this rapid spread is unusual. The morbidity varies considerably and depends on the strain of the virus and the inoculum delivered by the biting insect. Some horses become acutely ill and die after infection, whereas in others the infection is clinically inapparent. Outbreaks of disease associated with EIAV in horses of developed countries are rare. Animal Risk Factors Horses and ponies are susceptible to infection by EIAV and characteristically develop signs of the disease within days to weeks of infection. Mules also become infected and develop clinical signs similar to those of horses and ponies when infected with strains of the virus pathogenic to horses, but donkeys do not subsequently develop signs of the disease despite persistent infection with the horse-derived virus. The resistance of donkeys to horse-derived strains of EIAV is not definitive evidence that donkeys do not develop equine infectious anemia, and there is suspicion that strains of the virus pathogenic to donkeys exist. Methods of Transmission The source of all new infections with EIA is an infected horse, donkey, or mule. Horses are persistently infected, and clinically normal infected horses are a source of the virus. The virus can also be spread from clinically affected animals, which, because of the high concentration of virus in their blood, are a potent source of infection and important in the rapid spread of infection. Transmission of EIAV occurs almost exclusively through the transfer of contaminated blood or blood products. In field conditions this usually occurs through the mechanical transmission of contaminated blood from an infected horse to an uninfected horse by biting insects.

796

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Insect Vectors The insect vectors responsible for the transmission of EIAV between horses are all large biting flies, including Stomoxys calcitrans (stable fly), Chrysops spp. (deer fly), and Tabanus spp. (horse flies). Mosquitoes are not recognized as an important vector. Transmission is mechanical because the virus does not replicate in insects and is related to the large quantity of blood (10 nL) that the biting insects are capable of holding in their mouths. Infection occurs only if the feeding of the insect is interrupted. If this occurs the insect may attempt to feed again on the initial host or may seek another host that is close by. If the initial host is infected, the insect can carry blood from this animal to the second host and spread the infection. Tabanid flies can travel a distance of over 6 km, but when feeding is interrupted, the flies usually attempt to complete the meal on the initial host or a nearby animal and rarely travel more than 200 m to complete the meal. Insect factors that influence the likelihood of spread include the following: • Climate and season—tabanids prefer hot and humid conditions for feeding and breeding, and their activity is much reduced or absent in winter months. • Attractiveness of the host—foals are less likely to be bitten. • Proximity of hosts to woodlands— tabanids prefer treed or sheltered habitat. • Host housing—tabanids do not enter closed shelters. • Distance between horses—as noted earlier, tabanids prefer to complete an interrupted meal on the initial host or a nearby host. Other Means of Transmission Intrauterine infection can occur, although not invariably,24 and result in abortion or the birth of infected foals that often die within 2 months. Mares can be infected by insemination with semen containing the virus. Iatrogenic spread of infection can be important in some outbreaks. Infection can be readily achieved by the use of contaminated surgical instruments or needles or by the injection of minute quantities of virus, and the use of a common needle when injecting groups of horses can cause an outbreak of the disease. In enzootic areas, outbreaks have been caused by the use of untreated biological preparations of equine origin. This mode of spread is exemplified by the 2006 outbreak of the disease in Ireland.25,26 The virus was introduced in plasma illicitly imported into the country from Italy.25 The plasma was harvested from horses housed on a farm that had recently confirmed clinical cases of the disease. Foals were infected by transfusion of plasma, and mares then acquired the infection from their foals. The

route of transmission of infection from the foal to the mare is uncertain because it occurred during a period of low activity of potential insect vectors. Additional cases were related to hospitalization of horses with index cases.26 The only identified risk factor was duration of hospitalization, and there was no evidence of iatrogenic spread of the infection. There is the potential for aerosol spread of the virus in the close confines of the hospital. The virus is also capable of invasion through intact oral and nasal mucosae, wounds, and even unbroken skin, but these portals are probably of minor importance in field outbreaks. Transmission of infection from horse to horse seems possible via swabbing instruments used to collect saliva for doping tests. Economic Importance The difficulty of diagnosis and the persistence of the carrier state for periods of many years have resulted in embargoes on the introduction of horses into countries with a low prevalence of the disease, causing economic losses and interference with sporting events.

PATHOGENESIS Viral Multiplication After infection, EIAV multiplies in tissues that have abundant macrophages, with the spleen being the principal site of viral infection and propagation and accounting for over 90% of cellular viral burden. Viral replication occurs only in mature tissue macrophages, and circulating monocytes account for only 1% of the cellular viral burden. The concentration of cell free virus in blood, which can be as high as 10 TCID50% per mL, parallels the clinical course. Fever and other clinical signs develop within 2 to 7 days of infection as the concentration of virus in the blood increases; signs resolve as the viremia abates. There is a persistent but low-level viremia that persists for the life of the horse. The level of viremia in horses without clinical signs of the disease is very low and undetectable using conventional virus culture techniques but evident using PCR. The virus is detectable in low concentrations in most tissues of asymptomatic horses. During periods of relapse of the clinical disease the degree of viremia increases. On these occasions, the virus isolated from the blood has antigenic characteristics different from those of the virus that originally infected the horse. Antigenic drift of the gp45 and gp90 antigens, which occurs constantly even in asymptomatic horses with low levels of viremia, allows mutations of the virus, which then avoid immune surveillance, multiply, and cause clinical disease. The frequency of relapses of the clinical disease declines markedly after the first year of infection, and horses that survive become asymptomatic carriers.

Immune Reaction The immune response to EIAV is responsible for controlling replication of the virus and also plays an important role in the pathogenesis of the disease.8 The major clinical signs and lesions of equine infection anemia are attributable to the host response to the virus and not direct viral damage to tissue. Replication of EIAV stimulates a strong immune response that is detectable in horses and ponies within 7 to 10 days of infection. The initial infection is likely controlled by cytotoxic T-lymphocytes before the appearance of neutralizing antibodies. Antibodies to the p26 core protein are detectable by AGID test in almost all horses 45 days after infection; by 60 days after infection, antibodies to gp45 and gp90 are present. The neutralizing antibodies are specific to the phenotype of the virus causing the viremia, and this phenotype can change over time, as discussed earlier. Hypergammaglobulinemia develops. The immune response includes the production of virus-neutralizing antibodies, complement-fixing antibodies, and cytotoxic T-lymphocytes. The immune responses are responsible for the termination of viremia, although this effect is not mediated by antibody-dependent cellular cytotoxicity against EIAV-infected macrophages, but rather by development of neutralizing antibody and cytotoxic T-lymphocytes. The importance of neutralizing antibodies in control of the disease within an animal is indicated by the observation that viremia is never associated with a virus with a neutralizing phenotype already recognized by the horse. Most viruses in viremic horses consist of a complex of virus and antibody. The virus– antibody complex is readily phagocytosed by cells of the reticuloendothelial system, including tissue macrophages, and is involved in the development of the fever, depression, thrombocytopenia, anemia, and glomerulonephritis characteristic of the disease. Neurologic disease in horses with EIAV infection is attributable to viral infection of neural tissue but not necessarily neurons. Anemia and Thrombocytopenia The anemia characteristic of horses experiencing several febrile episodes of EIA is attributable to the shortened life of RBCs and decreased RBC production. Infection with EIAV shortens the life span of circulating RBCs to about 38 days, compared with the normal value of 130 days. The reduction in RBC life span is likely a result of the presence of virus–antibody complexes on the surface of RBCs with subsequent clearance of such cells by the reticuloendothelial system, as evidenced by the presence of sideroleukocytes in the peripheral blood of infected horses. EIAV also has a suppressive effect on erythroid series cells in bone marrow. Anemia occurs in Arabian foals with severe combined immunodeficiency infected with

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

EIAV, indicating that the anemia is not wholly a result of the adaptive immune response of the host. Anemia of chronic disease, which is in part a result of the limited availability of iron stores, likely contributes to the lack of bone-marrow response. Thrombocytopenia is a consistent feature of the acute, febrile episodes of EIA and has been attributed to the deposition of virus–antibody complexes on platelets, with subsequent removal of affected platelets by tissue macrophages. However, others have identified a primary production deficit resulting from an indirect, noncytocidal suppressive effect of EIAV on megakaryocytes. EIAV does not infect megakaryocytes, and the suppressive effect of infection is at least in part a result of tumor necrosis factors alpha and beta. Another explanation for the thrombocytopenia is increased removal of platelets because of increased in vivo activation and formation of platelet aggregates, a form of nonimmune-mediated platelet destruction. This was associated with increased thrombopoiesis and an increase in the proportion of young platelets in blood. The precise mechanisms underlying the thrombocytopenia associated with acute EIAV infection of horses is unclear. Platelets of EIAV-infected horses with clinical signs of disease have diminished function in vitro. Platelets from infected horses had greater amounts of fibrinogen bound to their surface, ultrastructural abnormalities consistent with activation, and diminished in vitro aggregation responses. Persistence of Infection The cell reservoir of the virus in persistently infected horses is unknown, as are the mechanisms underlying latency. However, the ability of retroviruses to splice a DNA copy of their genome into the genome of the host is probably important in the persistence of viral infection. The viral genome is detectable in clinically normal but persistently infected horses. Presence of viral DNA in host tissue is indicative of infection, whereas the presence of viral RNA in blood is suggestive of viral replication. This viral strategy allows the virus to escape immune surveillance of the host. Factors triggering a relapse of virus production from the latent genome are unknown, but relapse is associated with antigenic drift that enables the virus to evade host immune responses.8,27 Summary of Pathogenesis A likely scheme of pathogenetic events is as follows: • Primary entry and infection of tissue macrophages, especially in the spleen • Destruction of macrophages and release of virus and components • Production of antibodies to antigenic components

• Formation of antigen–antibody complexes, which induce fever, glomerulitis, anemia, thrombocytopenia, and complement depletion • Hemolysis or phagocytosis caused by specific complexes activating the reticuloendothelial system • Temporary iron-deficient erythropoiesis caused by delayed release of iron from macrophages • Subsidence of pathologic processes as virus-neutralizing antibody restrains viral multiplication in macrophages— the virus is incorporated into the host genome and becomes latent. • Appearance of a new antigenic variant of the virus and commencement of a new cycle of viral replication in macrophages and a new clinical episode—the antigenic variation results from changes in the surface glycoprotein of EIAV. • Less frequent recurrence of these episodes, with the horse becoming permanently asymptomatic—the animal can be said to have achieved an appropriate level of immune response sufficient to protect it against antigenic epitopes that are common to all EIAV strains.

CLINICAL FINDINGS

An incubation period of 2 to 4 weeks is usual in natural outbreaks of equine infectious anemia. Outbreaks usually follow a pattern of slow spread to susceptible horses after the introduction of an infected animal. On first exposure to infection, horses manifest signs of varying degrees, classified as acute or subacute. Occasionally the initial attack is mild to inapparent and may be followed by rapid clinical recovery. As a rule, there is initial anorexia, depression, profound weakness, and loss of condition. Ataxia, behavioral changes, hyperesthesia, and blindness occur, and in some horses is recorded as the only clinical abnormalities. There is intermittent fever (up to 41° C; 105° F), which may rise and fall rapidly, sometimes varying as much as 1° C (1.8° F) within 1 hour. Jaundice; edema of the ventral abdomen, prepuce, and legs; and petechial hemorrhages in the mucosae, especially under the tongue and in the conjunctivae, may be observed. Pallor of the mucosae does not occur in this early stage, and they tend to be congested and edematous. There is a characteristic increase in the rate and intensity of the heart sounds, which are greatly exacerbated by moderate exercise. Respiratory signs are not marked, and there is no dyspnea until the terminal stages, but there may be a thin serosanguineous nasal discharge. There is considerable enlargement of the spleen, which may be detectable per rectum. Pregnant mares may abort. Many animals recover from this acute stage after a course of 3 days to 3 weeks. Others

797

become progressively weak and recumbent, and they die after a course of 10 to 14 day of illness. Animals recovering from the acute disease may appear normal for 2 to 3 weeks and then relapse, with similar but usually less severe signs. Death may occur during such a relapse. Relapses continue to occur at intervals of as little as 2 weeks but usually cease after about a year. If they recur, they are usually associated with periods of stress and characterized by fever, increasing emaciation, weakness, ventral edema, and the development of pallor of the mucosae, a late sign of this disease. In this chronic stage, the appetite is usually good, although allotriophagia may be observed. Some affected animals appear to make a complete recovery, but they remain infected and can suffer relapses as many as 11 years later.28 Prolonged therapy with corticosteroids can cause such a relapse. Even in the absence of clinical illness, infected animals often perform less efficiently than the uninfected. Most deaths occur within a year of infection. Survivors persist as asymptomatic carriers.

CLINICAL PATHOLOGY

Hematological examination of horses with the acute disease reveals moderate to marked thrombocytopenia and anemia, which can be severe. Thrombocytopenia occurs during the initial episode, and can precede horses becoming serologically positive,25 and during relapses of the disease, is most severe during the febrile episodes, and can be sufficiently low that it allows petechial hemorrhages to develop. The anemia can become more severe with relapse (14% to 20%, 0.14 to 0.20 L/L) and is normocytic and normochromic. During the 2006 outbreak in Ireland, anemia was an inconsistent feature of the disease (40% of cases).25 The presence of sideroleukocytes (leukocytes containing hemosiderin) is considered highly suggestive of EIA. There are no characteristic changes in the white blood cell count, although presence of band neutrophils is common in the early stages of the clinical disease. Hypergammaglobulinemia may be present. Serum biochemical examination might reveal an increase in bilirubin concentration, a decrease in serum iron concentration, and increased glutamate hydrogenase (GLDH) activity in serum.25

DIAGNOSTIC CONFIRMATION

Diagnostic confirmation cannot be made based on hematologic or serum biochemical analysis and is achieved through detection of antibodies to the p26 core antigen of EIAV. Two tests are in general use: the AGID test (Coggin’s test), which is standardized using recombinant reagents,29–31 and a number of ELISAs, including a cELISA test.32–36 Results of AGID testing are available in 24 hours, whereas those of ELISA testing can be

798

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

available in as little as 1 hour. Commercially available ELISA tests detect antibody to the p26 antigen, antibody to the gp45 transmembrane protein, or both.37 The ELISA tests inherently have greater sensitivity (detect lower concentrations of antibody) than does the AGID, but often the characteristics of the commercial ELISA assays are modified to decrease the sensitivity (increase the lowest concentration of antibody detected by the kit) so that results obtained with these kits are concordant with those obtained by AGID.31 An advantage of an ELISA that detects antibodies to gp45 antigen is that, when combined with testing for antibodies to p26, the ability to detect infected horses with equivocal test results on a single test is increased. This is similar to the technique of using a Western blot test to demonstrate the presence of antibodies to more than one antigen, especially those against the gp45 and gp90 antigens, when equivocal AGID or ELISA results are obtained. False-negative reactions for either test occur because the horse lacks antibodies to the p26 antigen. The AGID and cELISA tests do not detect a recently infected horse that has yet to develop antibodies. Some horses do not develop anti-p26 antibodies for 45 days after infection. False-positive reactions can occur in foals born to infected mares. Colostral transfer of anti-p26 antibodies to the foal will be detectable up to 6 months after birth. Positive reactions to ELISA testing (to the p26 antigen) in samples that are negative by AGID testing can be the result of interspecies determinants. It is suspected that horses exposed to related lentiviruses produce antibodies to structural proteins that cross-react with the EIAV p26 antigen in ELISA, but not AGID, testing. An algorithm for testing of equine samples for EIAV is provided in Table 11-8. Tests to detect proviral DNA or viral RNA in blood and tissue have been developed and are useful in detecting the presence of virus when viral concentrations in blood and/or tissue are low. The identification of proviral DNA in the blood of infected horses is as specific and apparently more sensitive than the AGID in detecting infected horses. Experimental transmission of the disease to susceptible horses by the SC injection of 20 mL of whole blood or Seitz-filtered plasma is also used as a diagnostic test and is a valuable, although expensive and archaic, supplement to other tests. The donor blood should be collected during a febrile episode when the viremia is most pronounced, and the recipient animals should be checked for increases in body temperature twice daily. Molecular diagnostic tests offer the advantage of detection of viremia, but currently are limited because available primers for PCR do not reliably detect all strains of the virus.2,38

Table 11-8  Algorithm for testing horses for infection by equine infectious anemia virus37 Collect sample and separate serum from red cells as soon as possible. Tier 1 Test sample using ELISA. If negative, report the results. If positive, continue to Tier 2. Tier 2 Test with both same and at least one other ELISA format. If negative, report as negative. If positive in only one ELISA—report as negative. If positive in two or more ELISAs from different manufacturers, perform AGID   test. If negative, report as negative. If positive, forward to Tier 3. If inconclusive, forward to Tier 3. Tier 3 Test in all formats to confirm results. If confirmed, test by immunoblot. If gp90, gp45, and p26 positive (recognized), report as positive. If 2 major proteins are positive (recognized), report as positive. If only p26 is recognized, report as negative. AGID, agar gel immunodiffusion; ELISA, enzyme-linked immunosorbent assay.

• Serology—heart blood or pericardial fluid (AGID, ELISA) • Virology (if desired)—chilled spleen, liver, bone marrow, and perihepatic lymph node (ISO) DIFFERENTIAL DIAGNOSIS Acute disease Purpura hemorrhagica Babesiosis Equine granulocytic ehrlichiosis Equine viral arteritis Autoimmune hemolytic anemia Leptospirosis Parasitism Idiopathic thrombocytopenia. Chronic disease Internal abscessation (metastatic Streptococcus equi infection) Chronic inflammatory disease, neoplasia, and chronic hepatitis

TREATMENT No specific treatment is available. Supportive treatment, including blood transfusions and hematinic drugs, may facilitate clinical recovery, but it is important to remember that recovered horses are persistently infected and infectious for life.

CONTROL

NECROPSY FINDINGS In the acute stages, there may be subcutaneous edema, jaundice, and petechial or ecchymotic subserosal hemorrhages. There is considerable enlargement of the liver, spleen, and local lymph nodes. The bone marrow is reddened as a result of increased amounts of hematopoietic tissue and may contain focal infarcts. In the chronic stages, emaciation and pallor of tissues are often the only gross findings. Histologic examination is helpful in the diagnosis, even in asymptomatic chronic carriers. Characteristic lesions include hemosiderosis, perivascular infiltrates of round cells in many organs, and an extensive proliferation of the mononuclear phagocytic cells throughout the body. Glomerulitis, probably caused by the deposition of virus– antibody complexes on the glomerular epithelium, may be present. Lesions in the brain are a lymphohistiocytic periventricular leukoencephalitis. Lesions of interstitial pneumonia are common in horses with EIA.39 Culture of this virus is time-consuming, and the diagnosis is usually confirmed on the basis of a positive serologic test and typical microscopic lesions. Samples for Confirmation of Diagnosis • Histology—formalin-fixed spleen, liver, bone marrow, kidney, lung, heart

Control of EIA is based on identification and eradication or lifelong quarantine of infected animals, quarantine and testing of new stock, compulsory testing of imported horses, and efforts to prevent spread of the virus by controlling insect access to horses and use of strict hygiene when vaccinating or collecting blood samples from horses.40 The control of equine infectious anemia is still universally based on the eradication of the disease by identifying the infected, clinically normal animals with a serologic test and then destroying them. Identification is by means of the AGID or cELISA tests. The ability of a program of test and eradication to eliminate the disease is evidenced by the eradication of EIA from Hong Kong. An effective control program is described for Kentucky in the United States that permits the maintenance of infected horses with indelible identification and prescribed restrictions on housing. Control programs based on this test-andslaughter policy are under fire because of the view of horse owners that many asymptomatic horses, with very low infectivity, are being destroyed unnecessarily.41 A decision on the matter depends on whether the objective is eradication or containment, and if the latter, at what level. Until now the policy has been eradication, and it is obvious that another attitude is possible. Some flexibility in official attitude is desirable because of the

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

fallibility of the recommended control procedures and the devastating losses that can occur when the optimum environment for the spread of the disease is encountered. Foals from infected mares can be raised free of infection. Such foals have detectable antibodies to EIAV for up to 330 days on immunoblot and 260 days on ELISA testing because of transfer of maternal immunoglobulins in colostrum during the neonatal period. However, foals that are ultimately free of infection do not have detectable viral RNA in blood and have declining concentration of antibodies to EIAV. Foals should be isolated from infected horses as soon as feasible after diagnosis of EIAV infection in the dam. Restriction of introduction of infected horses into clean herds or areas is important to prevent introduction of the disease. Horses should be tested before introduction to the herd, and perhaps again in 1 to 2 months. If suspect horses are to be introduced, they should be kept under close surveillance for at least 6 months before being admitted. Horses known or suspected to be infected should be separated from all other uninfected horses, donkeys, and mules by a distance of at least 200 m. This recommendation is based on observations of the feeding behavior of tabanids, which are very unlikely to fly more than 100 m after an interrupted feeding. Suspect positive horses should be retested after at least 1month and probably at regular intervals thereafter. Operators of open stud farms and rest farms can also insist on proof of a negative test before admitting each horse. One deficiency of this policy is the long period of incubation of up to 45 days between infection and seroconversion to a positive test. In countries where the incidence is high, it is usual to control horse movement by a system of permits and certificates of freedom from the disease and to insist on skin branding or lip tattooing of all horses. Horses with a positive AGID or ELISA test should be allowed to move only under specified conditions. Draining of marshy areas and the control of biting insects may aid in limiting spread of the disease. A degree of protection may be obtained by the use of insect repellents and by stabling in screened stables. Great care must be taken to avoid transmission of the disease on surgical instruments and hypodermic needles, which can only be sterilized by boiling for 15 minutes or by autoclaving at 6.6-kg pressure for a similar period. Chemical disinfection of instruments and tattoo equipment requires their immersion for 10 minutes in one of the less corrosive phenolic disinfectants. All materials to be disinfected need to be cleaned of organic matter first. For personal disinfection, sodium hypochlorite, ethanol, or iodine compounds are safe, and for materials where organic matter is not removable, agents such

799

as chlorhexidine or phenolic compounds combined with a detergent are satisfactory. There are considerable problems associated with development of vaccines against EIA because only neutralizing antibodies are capable of causing sterile immunity and preventing infection. Neutralizing antibodies are specific for the homologous virus, but the large variation in phenotypes of the wild virus means that it will be difficult to stimulate neutralizing antibodies protective against all of the possible infecting phenotypes.42 Vaccines are available in parts of the world but are not in general use.43 Killed, whole-virus vaccines are safe, but subunit vaccines may actually enhance the occurrence of the disease. An experimental live attenuated EIAV DNA proviral vaccine affords complete protection in experimentally infected horses and has been widely used in China. The use of attenuated vaccines is associated with risk of reversion to virulence and lack of sterilizing immunity against heterologous virus challenge.38

37. Issel CJ, et al. Vet Rec. 2013;172:210. 38. Cook RF, et al. Vet Microbiol. 2013;167:181. 39. Bolfa P, et al. Vet Res. 2013;44. 40. Brangan P, et al. Equine Vet J. 2008;40:702. 41. Issel CJ, et al. Vet Clin Equine. 2014;30:561. 42. Craigo JK, et al. PLoS Pathog. 2015;11:e1004610. 43. Tagmyer TL, et al. J Virol. 2008;82:4052.

FURTHER READING

BABESIOSIS (TEXAS FEVER, REDWATER FEVER, CATTLE TICK FEVER)

Cook RF, et al. Equine infectious anemia and equine infectious anemia virus in 2013: a review. Vet Microbiol. 2013;167:181-204.

REFERENCES

1. Liu Q, et al. Viruses-Basel. 2015;7:3241. 2. Boldbaatar B, et al. J Virol Meth. 2013;189:41. 3. Capomaccio S, et al. Virus Res. 2012;163:656. 4. Dong J, et al. V et Microbiol. 2014;174:276. 5. Caij AB, et al. Transboundary Emerg Dis. 2014;61:464. 6. Dong J-B, et al. J Gen Virol. 2013;94:360. 7. Quinlivan M, et al. J Gen Virol. 2013;94:612. 8. Sponseller BA, et al. Virol. 2007;363:156. 9. Albayrak H, et al. Trop Animal Health Prod. 2010;42:1593. 10. Inci A, et al. Ankara Univ Vet Fak Der. 2013;60:281. 11. Marenzoni ML, et al. Turk J Vet Anim Sci. 2013;37:76. 12. Yilmaz O, et al. J Equine Vet Sci. 2013;33:1021. 13. Borges AMCM, et al. Res Vet Sci. 2013;95:76. 14. Cutolo AA, et al. Semina-Ciencias Agrarias. 2014;35:1377. 15. Gaiva e Silva L, et al. Rev Inst Med Trop Sao Paulo. 2014;56:487. 16. Guimaraes LA, et al. Rev Bras Med Vet. 2011;33:79. 17. Body M, et al. Pak Vet J. 2011;31:235. 18. Getachew M, et al. J Vet Med Animal Health. 2014;6:231. 19. Hussain MH, et al. Pak Vet J. 2012;32:247. 20. Jimenez D, et al. Open Vet J. 2014;4:107. 21. Lo C-H, et al. Taiwan Vet J. 2014;40:1. 22. Mangana-Vougiouka O, et al. Rev Scien Tech OIE. 2013;32:775. 23. Pagamjav O, et al. Microbiol Immunol. 2011;55:289. 24. Kuhar U, et al. Equine Vet J. 2014;46:386. 25. More SJ, et al. Equine Vet J. 2008;40:706. 26. More SJ, et al. Equine Vet J. 2008;40:709. 27. Schwartz EJ, et al. J Virol. 2015;89:6945. 28. Capomaccio S, et al. Vet Microbiol. 2012;157:320. 29. Alvarez I, et al. Clin Vaccine Immunol. 2007;14:1646. 30. Alvarez I, et al. Vet Microbiol. 2007;121:344. 31. Reis JKP, et al. J Virol Meth. 2012;180:62. 32. Alvarez I, et al. Rev Arg Microbiol. 2015;47:25. 33. Craigo JK, et al. J Virol Meth. 2012;185:221. 34. Hu Y, et al. Chin J Prev Vet Med. 2014;36:651. 35. Hu Z, et al. Appl Microbiol Biotech. 2014;98:9073. 36. Piza AST, et al. Prev Vet Med. 2007;78:239.

TICKS THAT TRANSMIT PROTOZOAN DISEASES Ticks are the most important vectors of many protozoan diseases, the protozoan in most instances surviving from generation to generation of ticks by infecting their eggs. Where control of these diseases is to be undertaken, it is necessary to know which ticks are vectors, how many hosts the tick parasitizes during a life cycle, and which animals can act as hosts. Much of the information on these points is fragmentary and only a summary is presented in Table 11-9. Additional information on the control of ticks is provided in Chapter 17.

Babesia spp. are a diverse group of tickborne, obligate, intraerythrocytic apicomplexan parasites infecting a wide variety of organisms.1–10 Infection of a vertebrate host is initiated by inoculation of sporozoites into the bloodstream while the tick takes a blood meal. Most babesial sporozoites directly invade circulating erythrocytes without a tissue stage of development. A few, notably, Babesia equi and Babesia microti, first invade lymphocytes, where they form motile merozoites, which then invade erythrocytes. Once erythrocyte invasion occurs, a seemingly perpetual cycle of asexual reproduction is established, despite a rapid development of a strong immune response. SYNOPSIS Etiology Babesia spp. Epidemiology Disease of tropical and subtropical countries. Occurs in cattle, sheep and goats, horses, cervids, and pigs. Transmission by blood-sucking ticks. Young calves have innate resistance. Endemic stability occurs in herds with a sufficient inoculation rate to immunize a high percentage of animals. Zoonotic implications Babesia bigemina and B. microti occurs in humans where suitable ticks are found. Human donor blood may be infected. Clinical signs Anemia, hemoglobinuria, jaundice, fever, high case-fatality rate. Clinical pathology Parasites in stained blood smear, positive serology. Polymerase chain reaction (PCR) for detection of parasite in blood.

Continued

Chapter 11 

800

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Necropsy lesions Thin, watery blood; pallor; jaundice.

Table 11-9  Ticks reported to transmit protozoan disease

Diagnostic confirmation Parasites in blood smear; vector present in environment.

Disease

Differential diagnosis list  A syndrome of acute hemolytic anemia should suggest the following alternative diagnoses:

Cattle

Protozoan

Vector ticks

Country

Babesia bigemina

Boophilus annulatus, B. microplus, B. (annulatus) calcaratus, B. decoloratus; Rhipicephalus appendiculatus, R. bursa, R. evertsi; Ixodes ricinus; Haemaphysalis punctata Ixodes persulcatus, I. ricinus; B. annulatus, B. microplus B. annulatus (calcaratus); Rhipicephalus bursa

North America, Australia, South America, Africa

Dermacentor sylvarum; Rhipicephalus bursa; Haemaphysalis punctata; Ixodes rinicus Rhipicephalus bursa; Haemaphysalis bispinosa; Haemaphysalis longicornis

Europe

Babesiosis

Cattle

Theileriosis Postparturient hemoglobinuria Bacillary hemoglobinuria S-methyl-L-cysteine-sulfoxide (SMCO) poisoning Leptospirosis

Babesia bovis

Treatment Diminazene aceturate and imidocarb. Control Tick control, vaccination with live vaccine, chemoprophylaxis with imidocarb.

Babesia berbera Sheep and goats

ETIOLOGY The nomenclature of these intraerythrocytic parasites is still subject to change; species in livestock include the following: • Cattle: species include Babesia bovis (B. argentina), B. bigemina, and B. divergens1,2 • Sheep and goats: B. motasi, B. ovis • Pigs: B. trautmanni, B. perroncitoi

Babesia motasi

Babesia ovis Babesia ovata Horses

Babesia caballi

Babesia equi

EPIDEMIOLOGY Geographic Occurrence The distribution of the causative protozoa is governed by the geographic and seasonal distribution of the insect vectors that transmit them (Table 11-10). Host Occurrence Bovine Babesiosis Babesiosis caused by B. bigemina and B. bovis is an important disease mainly in tropical and subtropical regions of the world.2,3 Both species are transmitted transovarially by Boophilus or Rhipicephalus ticks, but only tick larvae transmit B. bovis, whereas nymphs and adults transmit B. bigemina. Other species of Babesia rely on other ticks, including Haemaphysalis and Hyalomma. B. bigemina and B. bovis occur in the tropical and subtropical regions of Africa, America, Asia, Australia, and Europe (between 40°N and 32°S). B. divergens occurs in Europe and is the principal cause of babesiosis in the United Kingdom. Other species, such as B. divergens and B. major, occur in temperate regions. Bovine babesiosis is widespread in South Africa, for example, and the distribution of both B. bovis and B. bigemina is determined by the distribution of their tick vectors. The seroprevalence of B. bigemina in nonvaccinated cattle is a result of the high vectortick population and the endemically stable situation that can be achieved by a tickcontrol method that allows a reasonable

Pigs

Babesia trautmanni

Europe Former USSR Europe Iran Australia Africa

Former USSR India Japan

Hyalomma dromedarii; Africa Dermacentor (reticulata) marginatus, Former USSR and the Balkans, D. pictus, D. sylvarum; South America, Florida in the United States Hyalomma (excavatum) anatolicum, Africa, the Balkans, H. marginatum, H. volgense; South America, Rhipicephalus bursa, R. sanguineus Australia Hyalomma dromedarii; Rhipicephalus evertsi, R. sanguineus; Dermacentor marginatus, D. pictus; Hyalomma anatolicum, H. marginatum, H. uralense; Rhipicephalus bursa, R. sanguineus R. sanguineus (turanicus)

Former USSR

Theileriosis Cattle

Sheep

Theileria parva Rhipicephalus appendiculatus Theileria annulata Hyalomma anaticolicum Theileria sergenti Theileria mutans

Haemaphysalis sergenti Amblyoma variegatum; Haemaphysalis spp.

Theileria buffeli

Haemaphysalis spp.

Theileria ovis

Rhipicephalus bursa, R. evertsi; Hyalomma spp.; Rhipicephalus spp. Hyalomma anaticolicum

Theileria hirci

Africa Africa, Asia, former USSR, Europe, China, India Japan, Asia Africa, Asia Europe, former USSR, North America Australia Africa, Asia Europe Africa, Middle East Former USSR

number of ticks on cattle rather than relying entirely on intensive tick control and vaccination.1

babesiosis is of considerable economic importance in the areas infested with Rhipicephalus or Haemophysalis.

Sheep and Goats In sheep and goats, babesiosis is associated with species such as B. ovis and B. motasi and occurs in Africa, Asia, and Europe. Sheep

Porcine Babesiosis Associated with B. trautmanni and B. perroncitoi, porcine babesiosis occurs in Africa and Europe.

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

Table 11-10  Major Babesia species infective to domestic animals, their tick vectors, and their geographic distribution1 Domestic species affected

Babesia spp.

Major ixodid vectors

Known distribution

Babesia bigemina

Boophilus microplus Boophilus decoloratus Boophilus annulatus Boophilus geigyi Rhipicephalus everti

Africa, Asia, Australia, Central and South America, Southern Europe

Cattle, buffalo

Babesia bovis

Boophilus microplus

As for Babesia bigemina, but less widespread in Africa as a result of B. microplus competition with B. decoloratus

Cattle, buffalo

Babesia divergens

Ixodes ricinus

Northwestern Europe, Spain, Great Britain, Ireland

Cattle

Ixodes persulcatus Babesia major

Haemaphysalis punctata

Europe, northwestern Africa, Asia

Cattle

Babesia ovata

Haemaphysalis longicornis

Eastern Asia

Cattle

Babesia ovis

Rhipicephalus bursa

Southeastern Europe, northern Africa and Asia

Sheep and goat

Babesia motasi

Rhipicephalus bursa

Southeastern Europe, northern Africa and Asia

Sheep and goat

Babesia caballi

Dermacentor spp. Hyalomma marginatus Hyalomma truncatum Rhipicephalus evertsi

Africa, South and Central America, southern United States, Europe, Asia

Horses, donkey, mule

Babesia canis

Rhipicephalus sanguineus Dermacentor spp., Haemaphysalis spp. Hyalomma spp.

Southern Europe, North America, Asia, Africa, Australia

Dog

Babesia gibsoni

Haemaphysalis spp.,

Africa, Asia, Europe, North Dog America

Rhipicephalus sanguineus Babesia trautmanni

Rhipicephalus spp.

Equine Piroplasmosis In horses, donkeys, mules, and zebras, equine piroplasmosis is associated with B. caballi and “B. equi.”1,2 The latter parasite is now recognized as Theileria equi2 and will be retained within the context of piroplasmosis (disease caused by Babesia or Theileria) because the diseases caused by B. caballi and T. equi are similar clinically (see the section on equine piroplasmosis). Equine piroplasmosis occurs in much of southern Europe, Asia, and the Americas. For example, it is widespread in China and a cause for serious concern in northeastern China. In addition, equine piroplasmosis is also widespread in horses, mules, donkeys, and zebras in South Africa.2 Fortunately, Australia is free from equine piroplasmosis, but seropositive horses were temporarily imported into this country for the Sydney Olympic games in 2000. While in

Southern Europe, former USSR, Africa

Pig

Australia, seropositive horses were kept at particular restricted sites. Wildlife Babesiosis Among Babesia species that infect wildlife, B. odocoilei infects cervids, including the whitetailed deer (Odocoileus virginianus), American elk, and American woodland caribou (Rangifer tarandus caribou).1 Desert bighorn sheep (Ovis canadiensis nelsoni) and red deer (Cervus elaphus elaphus) are also susceptible to infection but do not exhibit clinical signs of disease. B. odocoilei is transmitted by ticks of the genus Ixodes. Various species of Babesia have been recorded in reindeer, including B. divergens and B. tarandirangiferis. Origin of Infection and Transmission Viable stages of Babesia are present in the bloodstream of animals in the active phase

801

of the infection. Ticks are the natural vectors of babesiosis; the causative parasites persist and pass through part of their life cycle in the invertebrate host. Both B. bovis and B. bigemina pass through part of their life cycle in the tick Rhipicephalus australis (previously Boophilus microplus). Other species of Boophilus and Rhipicephalus are major vectors of babesiosis, but other ticks of the genera Haemaphysalis, Hyalomma, Ixodes, and Dermacentor can be involved in transmission.1 Rhipicephalus australis is the main vector of babesiosis associated with B. bovis and B. bigemina in cattle production systems in Australia and Central and South America. Ixodes ricinus is a common carrier of B. divergens in the United Kingdom.1,2 Rhipicephalus and Haemaphysalis spp. are common ticks of sheep and/or goats.2 Knowledge of the life history of the tick is central to implementing effective control strategies against babesiosis or piroplasmosis. When adult animals become infected with Babesia, they can act as carriers for variable periods, up to 2 years. If they are constantly reinfected, as they are in an endemic environment, they can act as carriers for life. Ticks that parasitize only one host are easier to eradicate and cause less spread of the disease than those parasitizing multiple hosts. The control of ticks capable of surviving on both domestic and wild animals presents major challenges. Life Cycle and Development of Babesia The development of B. bovis and B. bigemina follows similar patterns. Babesia spp. do not parasitize any vertebrate host cell other than erythrocytes. Each sporozoite penetrates the erythrocyte membrane with the aid of a specialized structure called the apical complex. Once inside, it transforms into a trophozoite from which two merozoites develop by a process of merogony or schizogony (binary fission) (see Fig. 11-9). The tick becomes infected when ingesting erythrocytes containing piroplasms (gametocytes). They develop into male and female gametes in the tick gut. The microgametes fuse with macrogametes to produce motile zygotes. The zygotes then multiply, and the resultant “vermicules” invade numerous organs of the tick, including the ovaries. Therefore the infection passes through the ovary and the egg to the next tick generation (called transovarial transmission). Usually the female tick becomes infected, and sporogony takes place in the salivary glands of larval, nymphal, and/or adult ticks of the next generation. When the tick attaches to a new host, sporozoites mature. In B. bigemina, for example, some development occurs in the feeding larvae, but infective sporozoites take about 9 days to appear, and they therefore only occur in the nymphal and adult stages of the tick. Transmission can occur throughout the rest of the nymphal stage and by adult

802

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

transmitted in this way depends largely on the degree of parasitemia occurring for each species. Thus the probability of physical transmission is slight with B. bovis and higher with B. equi and B. bigemina.

LIFE CYCLE FOR BABESIA BIGEMINA

VetBooks.ir

IN CATTLE TICK ON HOST

Transmitted by nymph and adult stages

Salivary gland TICK ON GROUND

Sporogony in salivary glands

Sporozoite enters blood stream

Merogony (binary fission)

Schizogony in infected larvae

Polymorphic ray bodies Formation of ray bodies

Dormant in eggs

Egg laying

TRANSOVARIAL TRANSMISSION

Gamogeny

Engorged females

Aggregate of multinucleated ray bodies

Other organs

Fusion of gametes

Gametes

Zygote penetrates digestive cell

Ovaries Secondary schizogony (multiple fission)

Digestive cell

Kinetes Hemolymph

Primary schizogony

IN GUT OF ADULT TICK

Vcellogene cell (or basophilic cell)

Fig. 11-9  The developmental life cycle of Babesia bigemina in cattle and the ixodid tick vector Boophilus microplus. (Adapted from Mehlhorn H, Schein E. The piroplasms - life-cycle and sexual stages. Adv Parasitol. 1984;23:37-103; Gough JM, Jorgensen WK, Kemp DH. Development of tick gut forms of Babesia bigemina in vitro. J Eukaryot Microbiol. 1998;45:298306; Mackenstedt U, Gauer M, Fuchs P. et al. DNA measurements reveal differences in the life-cycles of Babesia-bigemina and B-canis, 2 typical members of the genus Babesia. Parasitol Res. 1995;81:595-604.)

females and males. For B. bovis, the formation of infective sporozoites usually occurs within 2 to 3 days of larval tick attachment. The host is infected with infected tick saliva. Particular species of Babesia can persist for several tick

generations, even in the absence of new infections. Contaminated needles and surgical instruments can transmit the infection physically. The ease with which infection can be

Immunity and Susceptibility to Infection The immune responses of cattle to infection with B. bovis or B. bigemina involve both innate and acquired mechanisms.4 The response directed against infections with Babesia is both humoral and cellular and is T-cell dependent. In addition, an age-related immunity to initial infection with B. bovis in cattle is well established, characterized by strong innate immunity in young calves. Mononuclear phagocytes are engaged as the primary effector cells on innate and primary immune responses, and nitric oxide has been identified as at least one babesiacidal molecule produced by activated mononuclear phagocytes. When B. bovis–infected erythrocytes grown in culture are exposed to nitric oxide, death of the parasites occurs rapidly within the erythrocyte. Innate Immune Mechanisms There is an age-related immunity to primary infection of cattle with B. bovis and B. bigemina. Young calves possess this strong innate immunity against B. bovis infection, which lasts for approximately 6 months after birth and is abrogated with the removal of the spleen.4 Interleukin IL-12 and IL-10 are important immunoregulatory cytokines. The protective innate response in young calves to infection with virulent B. bovis involves the early appearance of IL-12 and interferongamma transcripts in the spleen. This is followed by a short period of inducible nitric oxide synthase expression. In contrast, IL-12 and IFN-gamma (mRNA) expression in the spleens of adult cattle that died from infection was delayed and depressed, and it occurred within the context of IL-10 expression. Also, in contrast to calves, there was no detectable antibody response before death in adults. Acquired Immune Mechanisms Following B. bovis infection, antibodies directed against protective and nonprotective parasite antigens and host antigens are produced.4 Hyperimmune serum from cattle infected with B. bovis many times, or a mixture of IgG1 and IgG2 prepared from hyperimmune serum from cattle, can be used to passively immunize naïve calves against B. bovis infection, and the protection is specific. Splenectomized calves given hyperimmune serum and challenged with B. bovis recover as effectively as intact calves. Strong immunity occurs after natural infection with most Babesia spp. There appears to be little relationship between the degree of immunity and the level of

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

antibodies in serum. If the infection recurs repeatedly, the immunity is permanent. If the illness is treated rapidly and efficiently, and the protozoa are killed before antibodies are produced, no immunity occurs. If the infection is not repeated, the protozoa survive in the host for a variable time (e.g., 6 months), and then disappear. A sterile immunity persists for approximately 6 more months, and the host is susceptible again about 1 year after infection. These periods of latent infection and resistance/protection to reinfection are usually subject to significant variation and to different responses between breeds of cattle and species of Babesia. Despite the potential severity of the acute infection, individuals that survive generally develop immunity against disease, but not against infection, and could remain persistently infected. In the case of B. bovis, infection can persist for years, and even for the lifetime of the animal. Babesia infections have adapted well to survival in immune hosts. A number of phenomena are known to contribute to parasite survival: rapid antigenic variation, cytoadhesion and sequestration, binding of host proteins to the infected red blood surface, the monoallelic expression of different members of multigene families, and establishment of transient immunosuppression.4,5 The inoculation rate measures the daily probability of infection. This is based on the knowledge that animals exposed to the parasite in the first 9 months of life become infected, immune, and seropositive without showing any clinical signs of disease.1 Inoculation rates of 0.0005 and 0.005 are endemically unstable because a high percentage of animals will reach the age of 9 months without having been exposed to the hemoparasite. This results in a high risk of disease (endemic instability) because primary infections in older animals are usually severe and can be fatal. In situations of endemic instability, the vaccination of calves against hemoparasites can be used to ensure that the herd is immune. Cattle producers can use tick control to break the transmission cycle. Endemic stability is defined as the state in which the relationship between host, agent, vector, and environment is such that clinical disease occurs rarely or not at all.1 Endemic stability (herd immunity) in bovine babesiosis occurs when the rate of transmission (inoculation rate) of Babesia spp. by the tick vector is sufficient to immunize most susceptible calves before the loss of their resistance. In tropical areas with a large vector population, natural exposure usually occurs at an early age, and cattle are thus immune to subsequent challenges as adults. If at least 75% of calves are exposed to B. bovis infection by 6 to 9 months of age the disease incidence will be very low, and a state of natural endemic stability will exist.

Immunity.  Cattle develop a durable, longlasting immunity after a single infection with B. bovis, B. bigemina, or B. divergens. Different species of Babesia do not usually induce cross-protective immunity.4 Immunity to B. bovis and B. bigemina appears to last for at least 4 years. There is evidence that the presence of serum antibodies is not necessarily an indication of immunity. Conversely, the absence of detectable antibodies is not necessarily an indication of a lack of immunity.4 Risk Factors Host Factors B. indicus breeds of cattle are considerably more resistant to babesiosis than B. taurus breeds.1 This observation is thought to be a result of the evolutionary relationship among B. indicus, Boophilus/Rhipicephalus spp., and Babesia. Zebu and Afrikaner cattle have a higher resistance to B. bovis than British and European breeds; Santa Gertrudis and crossbred cattle occupy an intermediate position. Zebu-type cattle are also relatively free from the disease because of their resistance to heavy tick infestation. In Australia, B. bigemina is usually of lower pathogenicity than B. bovis and rarely lethal even when fully susceptible adult cattle are introduced to an endemic area. Inoculation studies with B. bigemina in Australia have shown that B. indicus and B. indicus cross-bred cattle are more resistant than the B. taurus cattle. Age Resistance.  In cattle, there is a variation in susceptibility to infection according to age, and the severity of babesiosis increases with age. Calves and foals from naïve dams are highly susceptible to infection and clinical illness from birth to 2 months of age, at which time they develop resistance that persists to approximately 6 months of age. Calves and foals from immune dams receive antibodies via the colostrum, and this passive immunity persists for 3 to 4 months after birth. The highest infection rate is in animals between 6 to 12 months of age; infection is uncommon in animals of greater than 5 years of age. Animals of less than 1 year of age are infected predominantly with B. bigemina and those of greater than 2 years of age by B. bovis. Calves of up to 1 year of age, although fully susceptible to infection, are resistant to disease. The average age at which calves in endemic areas become infected is 11 weeks (2 to 34 weeks), but clinical signs and pathologic changes are mild and relatively short-lived at this early age. After 6 months of age the number of infected animals in enzootic areas increases. In housed cattle, the level of serum antibodies in cattle are at their lowest when they come out of the barn in the spring, and this level gradually increases as they are exposed to infected ticks. In enzootic areas, cattle most commonly affected by clinical disease are those that are susceptible and introduced for breeding

803

purposes, intended for slaughter, or are “in transit.” Cattle indigenous to endemic areas are rarely affected because of the natural resistance of very young animals, and passive immunity via colostrum from immune dams is gradually replaced by a state of active immunity. Severe clinical cases occurring in cattle in such areas are usually caused by exposure to some stress, such as parturition, starvation, or intercurrent disease. Such breakdowns in immunity are most likely to occur if there is a superimposed infection with a different parasite, such as Anaplasma marginale. Environmental Factors There is seasonal variation in the prevalence of babesiosis, with the greatest incidence occurring soon after the peak of the tick population. For example, in England babesiosis mainly occurs in spring, summer, and autumn. Of the climatic factors, environmental temperature is the most important because of its effect on tick activity—higher temperatures increase activity. Humidity and rainfall have little effect, and even with temperature, the effect is limited once a threshold of 7° to 100° C (44° to 50° F) minimum temperature is exceeded. The heaviest losses occur in marginal areas where the tick population is highly variable in size, depending on the environmental conditions. In seasons when the tick population decreases, infection may die out, and immunity is lost. Under favorable conditions, when ticks multiply, infection can spread rapidly among susceptible individuals within a population or among/between populations. Comparable circumstances may be created artificially when an inefficient dipping or treatment program is used, which reduces the tick population to a low level and is subsequently unable to sustain control. Pathogen Factors Many intraerythrocytic hemoparasites survive host immune responses through antigenic drift or shift, which has been demonstrated for Babesia bovis.5 The molecular basis for antigenic variation in Babesia and its possible connection with cytoadherence and sequestration have been examined. There are different “strains” and antigenic variants of both B. bovis and B. bigemina. Babesia infections in cattle can relate to superinfection with antigenically distinct parasite populations. Antigenic change can provide Babesia with a temporary respite from attack by the host immune system and thus might prolong the infection period or cause disease relapses. Nonetheless, strain differences and antigenic variation do not appear to be of major importance in relation to the effect of a vaccine because cross-immunity between/ among strains of the same Babesia species usually provide adequate protection against one another.

804

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Economic Importance Bovine babesiosis is the most economically important of these diseases because of direct losses of production and because of the restriction of movement of cattle for trade by quarantine laws. Many animals die or undergo a long period of convalescence, resulting in major meat and milk production losses. Incidental costs of immunization and treatment add to the economic burden. With early, effective treatment, the mortality rate can be reduced to 5%. Zoonotic Implications Human cases of B. divergens infection have been reported in France, Britain, Ireland, Spain, Sweden, Switzerland, the former Yugoslavia, and the former USSR.1,6 Geographically, these cases coincide with B. divergens–infected cattle populations and areas in which Ixodes ricinus occurs, involving inhabitants of rural areas who are exposed to ticks by virtue of their occupation or their recreational activities. Most cases are reported between May and October, during the main season of tick activity. Cases of human babesiosis have been diagnosed sporadically, mainly in North America and Europe.6 Traditionally, cases of human babesiosis in Europe have been linked to B. divergens, whereas those in North America have been associated with B. microti. Autochthonous cases of B. microti infections have also been detected in Taiwan, Japan, and Europe. Recently, piroplasms similar to B. duncani and B. divergens have been implicated in human disease.6 In addition, B. venatorum (a B. divergens–like organism), which is probably a parasite of deer, was involved in the first documented cases of human babesiosis in Austria, Germany, and Italy. This evidence indicates that various Babesia species known to infect wildlife and domestic animals have the potential to cause human disease. Deerassociated zoonoses have become a particular public health concern, for instance, in the United States because human contact with deer ticks has increased as a result of the proliferation of deer, abandonment of farmland that reverts to thick secondary vegetation, and increased use of coastal sites for human recreation. This explains the increasing frequency of reported human cases of babesiosis, Lyme disease, and human granulocytic ehrlichiosis. Babesia spp. represent a potential threat to the blood supply for transfusion because asymptomatic infections in humans occur, and the spread of these hemoparasites via blood transfusions has been reported from various countries. Using the microaerophilous stationary-phase (MASP) culture technique, the parasites proliferate in a settled layer of blood cells. This provides the opportunity to examine the basic biology of the organism, host–microbe interactions, immune factors triggered by the parasite,

factors involved in innate resistance of young animals to infection, and antimicrobial susceptibility. Their in vitro cultivation might also be used to produce parasite antigens and attenuated strains of Babesia that could be used for immunization.

PATHOGENESIS

Babesia spp. are a diverse group of tickborne, obligate, intraerythrocytic apicomplexan parasites infecting a wide variety of organisms.2 Infection of a vertebrate host is initiated by inoculation of sporozoite stage into the bloodstream while the tick takes a blood meal. Most Babesia sporozoites directly invade circulating erythrocytes. Once erythrocyte invasion occurs, a perpetual cycle of asexual reproduction is established, despite the onset of a strong immune response.4 Acute Cases When an animal becomes infected, multiplication of the protozoa in the visceral (B. bovis) or peripheral (B. bigemina) vessels reaches a peak with the development of clinically detectable hemolysis, the principal pathogenic effect, after an incubation period of 7 to 20 days. The hemolysis results in profound anemia, jaundice, and hemoglobinuria. A fatal outcome as a result of anemic anoxia commonly follows. In surviving animals, there are ischemic changes in skeletal and heart muscles. In B. bovis infection, there is also profound vasodilation and hypotension, resulting from the stimulation of production of vasoactive substances and an associated increase in vascular permeability. Circulatory stasis and shock follow; disseminated intravascular coagulation (DIC) and subsequent fatal pulmonary thrombosis are also features. Cerebral babesiosis is possible. In contrast, B. bigemina is an uncomplicated hemolytic agent and does not exert these vascular and coagulation effects. Susceptibility to infection with Babesia spp. decreases with age, but the severity of disease increases. For example, calves of 5 to 6 months of age infected with B. bovis show limited clinical signs, cattle of 1 to 2 years of age have a moderately severe disease, and aged cows suffer a severe, often fatal disease. Intrauterine infection with B. bovis has been reported. Animals that survive become carriers, a state in which a subclinical infection is maintained by a delicate immunologic balance between protozoa and antibodies. This balance is readily disturbed by the stress of transport, deprivation of food, pregnancy, or intercurrent disease. Carrier animals are resistant to infection with B. bovis for up to 2 years. With constant reinfection, such as occurs in enzootic situations, protection is continuous. The ability of cattle to infect ticks is much longer (1 year) with B. bovis than B. bigemina

(4–7 weeks). Similarly, the peak incidence is at a younger age for B. bigemina, and the reinfection rate is more rapid. Some experiments have shown that merozoites can periodically disappear from peripheral blood in infected cattle. In pregnant cows, there is usually no apparent infection of the calf in utero, but passive immunity is transferred via colostrum to the newborn calf. Immunology Calves of less than 9 to 12 months of age are as susceptible as adult cattle to infection with B. divergens but are less likely to exhibit clinical disease. This phenomenon, known as inverse age resistance, is the result of an innate resistance in calves and is independent of the maternal immune status. Although offspring of resistant dams acquire specific antibodies (mainly IgG) via colostrum, these immunoglobulins are not required for protection because calves of susceptible dams without specific serum antibodies are equally resistant. Studies of B. bovis in vitro have shown that erythrocytes from very young calves were unfavorable for parasite development, possibly because of the inhibitory effect of fetal hemoglobin. Cattle that recover from acute infection with B. bigemina or B. bovis, either naturally or following chemotherapy, remain persistently infected and resistant to further disease following reinfection with the same strain.1,4 Immunization with dead piroplasms or extracts can induce protection against challenge with homologous or heterologous strains, indicated by low parasitemias and a diminished packed cell volume (PCV). Immunity does not last indefinitely, and in the absence of a reinfection, the animal becomes susceptible to reinfection. Specific immune responses include both cellular and humoral components. Monocytes and lymphocytes are the main agents of cellmediated immunity. Experimentally, the exposure of cattle to avirulent and virulent strains of B. bovis in a primary infection results in considerable antimicrobial activity in peripheral blood monocytes and neutrophils.1 This elevated antimicrobial activity coincides with the time that parasite numbers peak in the circulation and occurs before parasite clearance. This information suggests that peripheral blood monocytes and neutrophils are active mediators in the innate immune response to a primary infection with B. bovis. In cattle vaccinated against B. divergens, protection is associated with elevated mononuclear cell proliferation. In cattle infected with B. divergens, serum antibodies can be demonstrated even before infected erythrocytes appear in blood smears, suggesting that they have no inhibitory effect on merozoite replication. During secondary infection, protection seems to depend on the high specificity of some anti– B. divergens serum antibodies, rather than their level, because resistant animals often

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

have low levels of specific antibodies. The importance of the spleen in the specific immune response is indicated by the fact that the removal of the spleen following recovery may result in a clinical relapse. Specific serum antibodies produced against the parasites are used for serologic diagnosis. The highest titers are obtained in the sera of cows that have had a series of infections, but the degree of resultant immunity is not related to the specific antibody titer. The antibodies can be passively transferred via serum or colostrum. The immunity to different strain of B. bovis is specific. However, when an infection with a heterologous strain of the protozoa occurs, there is an increased immune response. As with cattle, B. ovis infection in sheep can produce an acute attack of clinical illness, parasitemia, and the subsequent development of immunity.

CLINICAL FINDINGS Cattle Babesia Bovis.  The acute disease generally runs a course of 3 to 7 days, and a fever of greater than 40° C (104° F) is usually present for several days before other signs emerge. This is followed by inappetence, depression, polypnea, weakness, and a reluctance to move. Hemoglobinuria is often present (known as “redwater” in some countries); urine is dark red to brown in color and produces a very stable froth. Anemia and jaundice develop, particularly in prolonged and severe cases. Diarrhea may occur. Muscle wasting, tremors, and recumbency develop in advanced cases, followed terminally by coma. Many severely affected animals die precipitately at this point, after an illness of only 24 hours. Metabolic acidosis can be present in a significant percentage of cases of bovine babesiosis. During the fever stage, pregnant cattle can abort, and bulls may become sterile for 6 to 8 weeks. Cerebral babesiosis is manifested by incoordination, followed by posterior paralysis or mania, convulsions, and coma. The mortality rate in such cases is high, in spite of chemotherapy. In those that survive, the febrile stage usually lasts for approximately a week, and the total course about 3 weeks. Cattle that survive recover gradually from the severe emaciation and anemia, which are inevitable sequelae. A subacute syndrome also occurs, particularly in young calves, in which fever is mild and hemoglobinuria is absent. The syndrome associated with B. divergens is similar to that of B. bovis, except that, in addition, there is spasm of the anal sphincter, causing the passage of feces with great force in a long, thin stream, even in the absence of diarrhea; this sign is referred to as “pipe-stem” feces. Babesia Bigemina.  Hemoglobinuria is present earlier and more consistently than in

B. bovis infection, and fever is less of a feature. Acutely affected animals are usually not as severely affected as those with B. bovis infection. There is no cerebral involvement, and recovery in nonfatal cases is usually rapid and complete. However, in some cases, disease can develop very rapidly, with sudden and severe anemia, jaundice, and death. Animals that recover from B. bigemina remain infective to ticks for 4 to 7 weeks and remain as carriers for only a few months. Sheep Anemia, fever, icterus, and hemoglobinuria are common. Wildlife Babesiosis of elk and caribou are characterized clinically by lethargy, hemoglobinuria, icterus, fever, recumbency, and sudden death.1 Elk infected with B. odocoilei may not have any clinical signs of disease but may become ill during periods of stress, such as during the rutting season, calving, transportation, or overcrowding. Other Species In all other species, the syndrome observed is clinically similar to that described for cattle.

CLINICAL PATHOLOGY Hematology Severe anemia with erythrocyte counts as low as 2 million/µL and hemoglobin levels down to 3g/dL occur in clinical cases in cattle, with anemia peaking 9 to 16 days following infection. Significant reduction in platelet numbers and a depression in the fibrinogen content of blood also occur. Demonstration of the Presence of Babesia Direct Examination of Blood Smears A diagnosis of babesiosis in clinically affected animals depends on the detection of piroplasms (merozoites) in Giemsa-stained smears of capillary blood; venous blood may give a false negative result for B. bovis infection. There is no exact correlation between the percentage of erythrocytes containing protozoa and the severity of the clinical signs. Also in B. bigemina infection, piroplasms are numerous in peripheral capillaries; B. bovis is less readily found. This difficulty can be largely overcome by using thick blood smears for detection. Microscopic examination can detect parasitemia of approximately 105 in thin blood films and 106 in thick blood smears. Therefore thick blood films are 10 times more sensitive and are more reliable for the detection of low-level B. bovis infection. Blood films should be prepared from capillary blood collected after pricking the tip of the tail or margin of the ear; blood from the general circulation may contain 20 times fewer B. bovis than capillary blood.

805

Transmission Test The inoculation of blood from a potentially infected bovine to susceptible splenectomized calves is a highly sensitive technique for the direct detection of Babesia. For this test, 50 to 100 mL of blood is injected into the recipient either SC or IV. In the latter case, the incubation period will be shorter. The recipient cattle are examined daily, and the blood is examined for protozoa at the peak of the febrile reaction. Carrier cattle infected with B. bovis and/ or B. bigemina are difficult to detect because of the small number of piroplasms in peripheral blood. Microscopic examination of stained blood smears is not a reliable technique for the detection of Babesia-carrier animals. The evaluation of the persistence of B. bovis and B. bigemina infections can be established by inoculating blood from donor cattle into splenectomized calves and measuring specific anti-Babesia serum antibody levels. Culture of Babesia Some Babesia spp. can be cultured in vitro.3 For instance, B. divergens from the blood of carrier cattle can be isolated using an in vitro culture technique in sheep erythrocytes. Babesia stages can be isolated 9 months after the acute babesiosis phase and can be successfully subcultured, cryopreserved, and resuscitated using culture medium. This culturing approach allows for detailed studies of the parasite. Preservation of live protozoa can be effected by cryopreservation, by culture in a medium containing infected bovine erythrocytes, and in simple culture media in special apparatus for long periods and in large numbers. Methods of Detection and Identification of Babesia spp. The accurate diagnosis of babesiosis is an important component of controlling babesiosis.7 Microscopy detection methods are inexpensive and rapid, but their sensitivity and specificity are limited. Improved methods are being developed, and they offer faster, more sensitive, and more specific options compared with conventional approaches. Methods based the detection of nucleic acids (DNA) and their amplification are the most sensitive and reliable techniques available today. For instance, PCR assays have been developed for the detection and identification of common pathogenic bovine, equine, and rodent piroplasms. Following specific amplification of the parasite DNA by nested PCR, the parasite species can be identified by PCR-coupled sequencing and/or fragment-length polymorphism.7 Other combined tests include ELISA using a recombinant B. bovis antigen, PCR, and a DNA probe, which can specifically detect even low-level infections. The DNA probe has the added advantage of being able

806

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

to detect protozoa in necropsy specimens and in tick tissues. The PCRs are most useful because of their high sensitivity and specificity, which makes them suitable for the detection of carrier animals. Serology Diagnosis of past or present infection can be demonstrated by any one of a wide range of serologic tests.7 Cattle Because of the difficulty in finding piroplasms in smears in animals during the subclinical stages of the disease, particularly in surveillance studies for the detection of the infection in herds or areas, much attention has been directed toward employing serologic tests.7 Such tests are well established, but most of them have limitations in terms of specificity and sensitivity. Moreover, it is not possible, on an individual animal basis, to distinguish between current and past infections, or between exposure and infection. Complement Fixation Test.  CFT is a commonly used serologic test for bovine babesiosis. Other tests assessed under field conditions include passive agglutination, indirect fluorescent antibody test (IFAT), indirect hemagglutination, ELISA, microplate enzyme immunoassay (EIA), latex agglutination, capillary agglutination, slide agglutination, and card agglutination. These tests have relatively good reputations, with EIA being particularly sensitive. Immunofluorescence Antibody Test.  IFAT has been a popular test used to distinguish between Babesia spp. and to demonstrate the presence of antibodies in a population of animals. IFAT differentiates between antibodies against B. divergens and other bovine babesias, but not between B. divergens and B. capreoli from red deer. ELISA.  An ELISA test using a crude antigenic preparation of B. bovis was assessed for the specific detection of IgM serum antibodies and achieved a specificity of 94% and sensitivity of 100%.1 Specific IgM antibodies against B. bovis have been reported to appear on the 11th day following inoculation in animals by R. australis ticks and on the 19th day after inoculation in of animals with infected blood. A competitive ELISA (cELISA) is another high-throughput method for detecting serum antibodies against hemoparasites.1 For instance, the gene encoding B. bovis rhoptry-associated protein 1 (RAP-1) was used to develop such an assay.1 This ELISA approach was reported to differentiate animals with B. bovis–specific antibodies from uninfected animals, and from animals with antibodies against other tick-borne hemoparasites, with high sensitivity and specificity (both ≥ 98.5%).

Sheep ELISA has been assessed for the detection of B. ovis in sheep. A latex agglutination test (LAT) using recombinant B. equi merozoite antigen 1 (EMA-1) was developed for the detection of antibodies to T. equi.1 It is a simple, rapid, relatively sensitive, specific, and inexpensive alternative to IFAT or ELISA.

NECROPSY FINDINGS

In acute cases of babesiosis in all species, in which patients die after a brief illness and during an anemic crisis, the typical findings are jaundice; thin, watery blood; pale tissues; enlargement of the spleen, which has a soft, pulpy consistency; and gross enlargement and dark-brown discoloration of the liver. The gallbladder is distended, with thick, granular bile; the kidneys are enlarged and dark; and the bladder contains red–brown urine. Ecchymotic hemorrhages are present under the epicardium and endocardium, and the pericardial sac contains an increased quantity of blood-stained fluid. In cattle, a characteristic manifestation is severe intravascular clotting. In subacute or chronic cases of relatively long duration, the carcass is emaciated but hemoglobinuria is absent; the other changes observed in acute cases are present but are less pronounced. The microscopic examination of blood smears taken from peripheral blood, from kidney and heart muscle, and, in the case of suspected B. bovis infection, from the brain, is mandatory for clinching the diagnosis. Smears from blood and most tissues must be made within 8 hours of death, within 28 hours for the brain, and stained with Giemsa for the detection of B. bovis. Direct fluorescent antibody staining of smears permits the use of slightly “older” tissues. Organ smears are still usable 5 days after collection, provided that they are kept at 22° C (72° F). The morphology of B. bigemina changes quickly after the host’s death, so that zoites resemble those of B. bovis. Blood collected after death can also be used for detection of serum antibodies in serologic tests. DIFFERENTIAL DIAGNOSIS Preferably, the presence of the tick vector should be collected and verified before a definitive diagnosis of babesiosis can be made, unless an animal has left a known enzootic area within the preceding month. Clinically, a high morbidity and case-fatality rate in cases displaying jaundice with hemoglobinuria and fever are suggestive, but confirmation of the diagnosis by microscopic examination of stained blood smears, complementary immunologic or molecular tools, and/or by transmission experiments is required. A necropsied animal with

splenomegaly, jaundice, hemoglobinuria, swollen and dark kidneys and liver, and/or myocardial ecchymoses is suggestive of babesiosis/piroplasmosis, but the diagnosis needs to be confirmed by traditional or molecular laboratory testing of tissues for the presence of the parasite stages (merozoites/ piroplasms). Differential diagnosis list A syndrome of acute hemolytic anemia should suggest the following alternative diagnoses: Cattle (see Table 11-11):

Theileriosis (caused by Theileria)—very similar clinically and differentiable only based on laboratory examination Postparturient hemoglobinuria—does not require the presence of vectors, occurs only in recently calved cows in full lactation and on low-phosphorus diets, and is characterized by the absence of protozoa from blood and tissues Bacterial hemoglobinuria—characterized by a necrotic infarct under the diaphragmatic surface of the liver in cattle grazing lush pasture S-methyl-L-cysteine-sulfoxide (SMCO) poisoning—occurs only in cattle grazing crops of rape or other Brassica spp. Leptospirosis—occurs only in this form of the disease in calves kept in unsanitary conditions that are wet underfoot. Diagnosis of this disease depends on isolation of the leptospires.

TREATMENT Primary treatment is aimed at killing the parasite(s) in the patient.7,8 Effective drugs are available for use in cattle, but the initial phase of the disease is acute; if treatment is delayed for too long, the animal may succumb to the anemia, in spite of chemotherapy. If the illness is a consequence of vaccination with live vaccine, care must be taken to avoid a complete sterilization of the blood before sufficient serum antibody is produced against the parasite(s) to elicit a durable immunity. Treatment has no suppressive effect on the protozoa that are residing in the ticks parasitizing the cattle at the time. Drugs such as diminazene aceturate, imidocarb dipropionate, amicarbalide diisethionate, and phenamidine have been used against Babesia. Parvaquone, buparvaquone, and alovaquone are introductions with good reputations from clinical trials. Tetracyclines have been used extensively, but their use in acutely sick animals has been discontinued. There is some use in the simultaneous administration of live Babesia in a chemosterilant situation; the parasite is controlled and effective immunization is achieved. Cattle For many years, three babesicides, quinuronium sulfate (and generics), amicarbalide

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

807

VetBooks.ir

Table 11-11  Differential diagnosis of diseases of cattle in which red urine is a principal manifestation CLINICAL AND LABORATORY FINDINGS

Disease

Epidemiology

General

Urinary

Clinical pathology

Diseases with hematuria Enzootic Subjects older than 1 year. Persistent intermittent hematuria; Persistent, hematuria Endemic to specific areas with hemorrhagic anemia, acute or chronic. intermittent access to bracken. Rectal in acute cases nil; chronic cases have hematuria. local or diffuse thickening. Long course, death by anemia. Enzootic bovine pyelonephritis

Adults only. Sporadic cases usually. May be a series suggesting origin in one bull and relationship to mating events.

Mild fever. Frequent painful urination, Intermittent toxemia. Late cases, rectal examination hematuria and shows cystitis, ureters thickened and pyuria. enlarged, kidneys the same. Pain on palpation. Long course, death by uremia.

Diseases with hemoglobinuria Babesiosis (B. Outbreaks in marginal areas in High fever, pallor, severe jaundice heavy tick seasons in calves. terminally. bigemina and B. Incubation 2–3 weeks. 90% bovis) morbidity and mortality.

Urine has no pus, eukocytes, or bacteria.

Urine has pus, erythrocytes, eukocytes, C. renale on culture catheter sample.

Red urine, hemoglobinuria.

Babesia in red cells in smear. Transmission test. Many serologic tests.

Tropical theileriosis Transmitted only by ticks of (Theileria Hyalomma spp. annulata)

Fever, anorexia, lymph node enlargement.

Hemoglobinuria.

Piroplasms in red cells; schizonts in lymphocytes from liver biopsy. Serologic tests. Hyalomma spp.

Postparturient hemoglobinuria

Postcalving 2–4 weeks. Adult dairy cows in 3rd-6th lactation. Sporadic but tends to endemicity on individual farms. Lowphosphorus or low-copper diet.

Acute onset, weakness, tremor, pallor, bounding pulse, loud heart sounds, tachycardia. No jaundice. Mortality 50%. Long convalescence. Die of anemia, especially if stressed.

Deep brown to black frothy.

No cells in urine but good deposit on standing. Severe hemolytic anemia. Serum inorganic P < 1.5 mg/dL and down to 0.1 mg/dL.

Bacillary hemoglobinuria

Summer on irrigated pasture. Often found dead. Very acute onset, Sporadic. Very few cases. hemolytic anemia plus toxemia. Fever Endemic to particular farms. 41° C (105° F). Abdominal pain, pain on Mortality 100%. percussion right anterior abdomen. Diarrhea. Shallow, rapid respiration as a result of diaphragmatic pain.

Deep red brown, no cells.

Hemolytic anemia, increased serum bilirubin.

Leptospirosis (L. interrogans Pomona only, not L. hardjo)

Calves high mortality 50%. Hemolytic disease mostly in young calves. Adults low mortality < 5%. Sudden-onset septicemia with red urine. Abortion storm more common Severe toxemia, fever 40.5–41.5° C in adults. Many subclinical (104.5–106° F). infections in adults. Mucosal petechiae, pallor, and jaundice. Adults have thick orange milk all quarters.

Red urine, hemoglobinuria.

Initially leptospiruria 3 days. Leptospiruria by intraperitoneal injection into guinea pigs. Rising titer leptospira antibodies, with peak 4 weeks after infection.

Chronic copper poisoning

Rarely if at pasture. Copper supplement in a swine diet by mistake.

Hemoglobinuria, High liver copper on biopsy some 2000 ppm dry material. High methemoglobinuria plasma ceruloplasmin and copper.

isethionate, and diminazene aceturate, were available in most countries for the treatment of bovine babesiosis. In the 1970s, imidodocarb dipropionate was introduced, and it became the drug of choice in countries that licensed it, because in addition to its therapeutic utility, it also proved to be an effective prophylactic at twice the therapeutic dose. Currently, it is the only babesicide on the market in most countries of Europe. Quinuronium sulfate and amicarbilide have been withdrawn because of manufacturing safety issues; diminazene, which is widely used in

Sudden onset, weakness, pallor, jaundice, death usually in 24–48 hours.

the tropics as both a babesicide and a trypanocide, also was withdrawn in Europe for marketing reasons. Imidocarb is most toxic when given IV; IM or SC administration is recommended. Side effects of this drug include coughing, muscular tremors, salivation, colic, and local irritation at the site of injection, following the administration of high doses. Although it is regarded as being slower in action than quinuronium sulfate, it is the only babesicide that consistently clears the host of parasites. In the past, the persistence of small numbers

of parasites in the host was deemed necessary for the maintenance of resistance to reinfection. However, the concept of premunition appears no longer to be accepted. Premunition is used to describe resistance that is established after the primary infection has become chronic and is only effective if the parasite persists in the host. It was thought that only cattle actually infected with Babesia were resistant to clinical disease. If all organisms were removed from an animal, resistance was thought to wane immediately. However, cattle apparently cured of Babesia

808

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

infection by chemotherapy are resistant to challenge with the homologous strain of that organism for several years. The presence of infection does appear to be mandatory for protection against heterologous strains. Although a certain period of antigenic exposure is necessary before treatment to facilitate the establishment of immunity, cattle treated with imidocarb dipropionate ultimately have a solid immunity. Long-term persistence of low-level parasitemia is now considered a disadvantage. Remaining parasites may give rise to recrudescence under adverse conditions, treated cattle may act as a source of infection, and parasites surviving low levels of babesicide may acquire resistance. Imidocarb provides “protection” from clinical disease for 3 to 6 weeks, but allows a sufficient level of infection for immunity to develop. This strategy is highly effective if the host is assured to be exposed to babesiosis during the period of protection, either through a tick bite in geographic areas where babesiosis is endemic or by inoculation of live parasites. Acquired immunity then takes over from “drug protection,” and the animal passes smoothly to a resistant state without an intermediate clinical stage. However, if infection rates are sporadic or if very high doses of imidocarb are used, a complete inhibition of parasite development will hinder the mounting of an adequate immune response. The major issue associated with this approach is concern regarding drug residues in milk and meat, which has led to the withdrawal of imidocarb in several European countries.2 Imidocarb (Imizol) This and the related drug, amidocarb, are effective babesicides for cattle at the dose rate of 1 mg/kg BW. At 2 mg/kg BW, it eliminates the parasites from the host and maintains some residual activity; noninfected cattle derive a month’s resistance to clinical infection but can be infected subclinically. Therefore, imidocarb can be used to “protect” cattle when vaccination is undesirable (e.g., during pregnancy) or when exposure to infection is short-lived. Conversely, it can be used to temporarily protect animals before vaccination. The drug can be given SC. The hydrochloride form is inclined to be an irritant; propionate is less irritating. Sheep Diminazene aceturate is effective as a treatment in sheep (3.5 mg/kg BW on two successive days, or 12 mg/kg BW as a single dose). Supportive Treatment In all species, treatment regimens for severely affected sheep should include blood transfusions and antishock preparations. In chronic cases and convalescent patients, hematinics should be provided.

CONTROL (BOVINE BABESIOSIS) Prevention and Biosecurity Preventing the introduction of the disease into a nonenzootic area depends on effective quarantine to prevent the introduction of the vector tick and laboratory testing to ensure freedom of the importee from infection with the pathogen(s).1 Control The control of bovine babesiosis in an area depends on the control the tick vector. Eradication is usually not achievable/practical because of the high cost to local wildlife, some of which can be hosts to ticks. Other challenges encountered include the following: • The difficulty of getting a complete muster of all cattle on every dipping day • Multihost ticks, which can be infective but temporarily not resident on an animal on dipping day • The spread of ticks or infested cattle as a result of environmental issues, such as floods or windstorms • Illegal movement of cattle without a permit Other issues include the persistence of Babesia through successive generations of the tick vector and the resistance of ticks to acaricides, which is also a factor relating to the infestation level of cattle. The effect of different tick (Rhipicephalus microplus) control strategies (none, threshold, and strategic) on endemic stability and the likelihood of babesiosis (Babesia bovis) has been examined in parts of South America using a computer simulation model based on weekly tick counts. The cattle population was in a state of enzootic stability, with an inoculation rate exceeding 0.005 throughout the year. Threshold dipping strategies did not increase the risk of babesiosis. Strategic dipping resulted in an extended period of enzootic instability lasting 30 weeks, which required protection of the herd by vaccination. Therefore strategic dipping is proposed to lead to effective control or eradication of Babesia from tick and cattle populations, but it would not result in an eradication of the tick vector. This situation could lead to subsequent outbreaks if Babesia carrier animals were introduced into the herd. Strategic tick control could be accompanied by concurrent vaccination against babesiosis. Limitation of Prevalence To limit prevalence at economically sustainable levels requires different solutions in different circumstances. It is largely dependent on tick control through frequent application of acaricides, chemotherapy to kill Babesia in the cattle host, and, to a lesser degree, by immunization of cattle.7–9 These measures are only partly effective and are time-consuming and expensive. The reason for the poor performance of vaccination procedures, even after a great deal of research, is

that the mechanisms of immunity against protozoa, particularly Babesia spp., have not been explored in great detail. Further investigations need to elucidate how immune responses to these parasites work. Aspects that need to be considered to limit prevalence are the following: • Susceptible cattle moving into an enzootic area need prior vaccination. • Marginal areas abutting enzootic areas in which tick populations vary with climatic change, so that resident cattle lose immunity after some dry years and are then exposed to infection when wet years. foster an increased prevalence of ticks in these areas. Vaccination before outbreaks are predicted to commence is recommended, if forecasting is possible, and temporary chemoprophylaxis after outbreaks have commenced. • In enzootic areas where losses are occurring as a result of environmental stress or, particularly, concurrent infection with another pathogen (e.g., Anaplasma marginale), or where the tick population has been decimated by overzealous dipping, chemoprophylaxis and relaxation of dipping are recommended. Exposure of cattle to ticks is important to ensure to maintain a state of infection and immunity. Vaccination Vaccination with live and dead whole parasites, crude parasite extracts, and isolated parasite antigens has been used, with varying degrees of success.9 Several findings support the development of vaccines against babesiosis. First, cattle that recover from a primary Babesia infection or that have been immunized with attenuated parasites are resistant to challenge infection. Second, the immunization of cattle with native Babesia antigen extracts or culture-derived supernatants containing secreted Babesia antigens elicit protective immunity against both homologous and heterologous challenge. The characteristics of cattle farms on which the exposure of young cattle to tick fever organisms is sufficient to ensure that immunity is high and the risk of clinical disease is low (endemic stability) can be compared with those farms on which exposure is insufficient (endemic instability) to study and understand the relationships between the management of ticks and tick fever. In Queensland, Australia, for example, many cattle herds do not have sufficient exposure to B. bovis, B. bigemina, or A. marginale to confer endemic stability for tick fever.1 For B. bovis, the major cause of outbreaks of clinical disease in Queensland, less than half of the herds had evidence of endemic stability. The decision to leave some ticks on cattle, in an effort to induce endemic stability, did increase the likelihood of endemic stability to A. marginale. However, it was ineffective, because only 26% of herds

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

had endemic stability against all three pathogens. Thus, given the low proportion of herds with endemic stability to tick fever organisms and the high likelihood of clinical disease, vaccination is recommended to protect dairy cattle from tick fever throughout tick-infested areas.

of Babesia, or from parasites grown in vitro.9 The vaccines are provided either chilled or cyropreserved. In spite of the costly and time-consuming nature of producing such live vaccines, they have usually provided greater than 95% protection for the life of the cattle vaccinated.

Live Vaccines Vaccines incorporating live, attenuated strains of B. bovis and B. bigemina have been used routinely or experimentally in Australia and a number other countries.9 The literature on designing blood-stage vaccines against B. bovis and B. bigemina. has been reviewed.9 The data available on the efficacy, degree, and duration of immunity elicited by live vaccines against B. bovis and B. bigemina infections in Australia have also been reviewed.9 Most of the available live vaccines have been produced in government-supported production facilities in Australia, parts of South America, South Africa, and Israel. These vaccines include bovine erythrocytes infected with selected strains of the parasites. The risk of contamination of blood-derived vaccines is a concern and makes postproduction quality control essential; unfortunately, such quality control is beyond the means of some endemic countries. Techniques developed in Australia over many decades have formed the basis for the production of live Babesia vaccines in most countries where they are used. There is no reliable evidence to indicate that current live vaccines might spread disease from vaccinated to unvaccinated cattle.

Frozen Vaccine.  Frozen vaccine is superior to chilled vaccine because of its long shelflife, which allows postproduction testing of potency and safety before dispatch.9 Glycerol is used as cryoprotectant in Australia and is preferred over dimethyl sulphoxide because it allows postthaw storage life of the vaccine for at least 8 hours (at temperatures of 4° to 30° C (39° to 86° F)). Frozen vaccines are transported in suitably insulated containers with liquid N2 or solid CO2, which limits the ability to supply vaccines to all destinations. To ensure infectivity, the vaccine must be used within 8 hours of thawing, and once thawed should not be refrozen. A frozen bivalent B. bovis and B. bigemina vaccine and frozen monovalent B. bovis and B. bigemina vaccines using dimethyl sulphoxide as the cryoprotectant are produced in some countries. If dimethyl-sulphoxide is used, a vaccine should be used within 30 minutes of thawing.

Origin and Purification of Strains.  Since 1990, three strains of B. bovis and one of B. bigemina (G strain) have been used to produce vaccines in Australia. After testing for virulence, immunogenicity, and purity, suitable strains are preserved as master stabilates in liquid nitrogen. Attenuation of Parasites Babesia Bovis.  The most reliable method of reducing the virulence of B. bovis is the rapid passage of strains through susceptible, splenectomized calves. Attenuation usually occurs after 8 to 20 calf passages. Babesia Bigemina.  Rapid passage in splenectomized caves is not reliable, but the virulence of B. bigemina decreases during prolonged residence in latently infected animals. A single B. bigemina isolate (G strains) has been used in the Australian and South African vaccines for many years. Vaccine Specifications Live vaccines have proven very effective and reasonably safe, particularly when vaccination is restricted to cattle of less than 1 year of age, when they still are resistant to disease. Vaccines are derived from splenectomized donor calves infected with attenuated strains

Chilled Vaccine.  Most of the babesiosis vaccines produced to date have been provided in a chilled form.9 In Australia, 35 million doses were supplied between 1996 and 2003. It is popular because of ease of production, ease of transportation even with limited resources, ease of use, and low cost. The chilled vaccines used in Australia contained 1 × 107 B. bovis, 2.5 × 106 B. bigemina, and 1 × 107 Anaplasma centrale organisms per 2 mL dose. A chilled vaccine has a very short shelf-life of approximately 4 days, which requires rapid, reliable means of communication and transportation to ensure viability. Chilled vaccines can remain viable for up to a week if stored at 4° C (39° F). To reduce the risk of neonatal hemolytic disease in calves (alloimmune hemolytic anemia) of vaccinated dams, the vaccine should not be used repeatedly. Most owners vaccinate only young animals, seldom more than twice. A reduction of the vaccine dose and the use of a cell-free diluent have essentially eliminated the problem in Australia. The development of effective live vaccines against bovine babesiosis in Australia required laboratory and field research over the period from 1959 to 1996, and it is a remarkable success story of veterinary medicine.9 The most significant change occurred in 1964 with the traditionally used carriers of Babesia being replaced as vaccine donors by acutely infected splenectomized calves. This ensured that the infectivity of the vaccine and was fortuitously associated with a reduction in the virulence of the B. bovis

809

vaccine. The vaccine reduced serious losses from babesiosis in vaccinated cattle in Australia to very low levels and gained acceptance worldwide. The demand for a live, trivalent “tick fever” vaccine containing B. bovis, B. bigemina, and A. centrale produced by the Department of Primary Industries in Queensland, Australia, increased from less than 10,000 doses in 1988 to 500,000 doses in 2001.1 The challenge to obtain B. bigemina parasitized erythrocytes on a large enough scale from infected splenectomized calves to meet the demand was achieved by reducing the dose rate of infected cells without affecting immunogenicity and still leaving a safety margin of at least 50-fold for infectivity. This change quadrupled the potential yield of doses per calf and allowed the department to meet the increased demand for the B. bigemina vaccine. Use of Live Vaccine Cattle Born in Tick-Infested Regions.  Any factor affecting the survival of the tick vectors will affect the risk of babesiosis. An increased number of ticks will increase the threat of disease until an endemically stable situation develops. Conversely, reduced tick numbers will increase the longer-term risk of babesiosis because of the reduced natural exposure of calves. Therefore cattle owners in endemic areas in Australia are advised to supplement natural exposure by vaccinating calves at weaning age. Vaccination is also recommended if cattle are being moved within an endemic area. Susceptible Cattle Imported Into VectorInfested Country or Region.  Large numbers of cattle, predominantly B. taurus, are being imported into tropical developing countries to upgrade local livestock industries. This has resulted in significant economic losses from tick-borne diseases, including babesiosis. Vaccination of naïve cattle moving from tick-free to endemic areas within Australia is usually very effective. This practice has played a crucial role in making the livestock industries in these countries more sustainable and competitive in meeting market demand with regard to breed. The K strain of B. bovis and G strain of B. bigemina from Australia have been shown experimentally to be protective in South Africa and Sri Lanka. Vaccines containing these strains have also been used with beneficial results in parts of Africa, South America, Malaysia, and the Philippines. Control of Outbreaks.  Use of a vaccine in the face of an outbreak is common practice in Australia. Superimposing vaccination on a natural infection will not exacerbate the disease but will preempt the development of virulent infections in a proportion of the herd not yet exposed to the pathogens. To

810

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

prevent further exposure, the group should also be treated with an acaricide capable of preventing tick attachment from the time of diagnosis to 3 weeks after vaccination. Injectable or pour-on formulations of ivermectin and moxidectin and fluazuron are highly effective acaricides but do not prevent the transmission of Babesia. Clinically affected cattle should be treated as soon as possible with a suitable babesiacide. In the case of a severe outbreak, it might be advisable to treat all the cattle with a prophylactic compound, such as imidocarb or diminazene, and to vaccinate them later when the drug residue will not affect the replication of the vaccine parasite(s). Hazards and Precautions of Live Vaccine Use Severe Reactions.  The likelihood of vaccine-induced reactions has been reduced with the development of attenuated strains. However, there is always a risk of such reactions when highly susceptible adult cattle are vaccinated. Calves of 3 to 9 months of age have a high level of natural resistance and a low risk of reactions. In some countries, vaccination is only recommended for calves, whereas in Australia and South Africa, adult cattle can be vaccinated, provided proper precautions are taken. Concurrent infections may increase the likelihood of reactions. In pregnant cows, fever associated with reactions has the potential to cause abortion; in large bulls, there can be a temporary loss of fertility. In the case of valuable cows and bulls, their body temperatures should be monitored if vaccine-induced reactions occur, and those with prolonged fever should be treated with a babesiacide. Lack of Protection.  Since the introduction of a standardized method of production in Australia, live antibabesiosis vaccines have been highly effective. In most cases, a single vaccination provided lasting, probably lifelong immunity against field infections with antigenically distinct strains. However, some failures have occurred and are thought to have been associated with a loss of immunogenicity because of frequent passaging of the vaccine strains in splenectomized calves. This was overcome by replacing the vaccine strain. To prevent future recurrences of vaccination failure, the number of passages of vaccine strains of B. bovis is limited by frequently reverting to a master stabilate with a low passage number. Other failures may be associated with the immune responsiveness of the host and the immunogenicity of vaccine strain subpopulations. A single inoculation of cattle at 6 to 9 months of age with an attenuated vaccine containing B. bovis and B. bigemina usually provides good, long-lasting protection. At this age, the risk of vaccine reactions is minimal. Immunity following use of a live B. bovis vaccine lasts for at least 4 years,

possibly less for B. bigemina. It is known to persist even after elimination of Babesia infections, and studies on drug-cured cattle suggest that the degree of acquired immunity relates to the degree of antigenic stimulation (duration of prior infection) rather than the presence of live parasites. There is no evidence of a loss of immunity with time, and revaccination is mostly not required. Revaccination is advisable when there is uncertainty over the reliability of previous procedures, to ensure that all animals seroconvert, or if there has been a change in the strains used in the vaccine. A cryopreserved vaccine containing in vitro culture-derived (attenuated) stains of B. bovis and B. bigemina can achieve protection in 90% of vaccinated cattle against the virulent field strains of Babesia.1,9 Inherent disadvantages of vaccines derived from the blood of animals include the risk of reactions or contamination with pathogenic organisms, sensitization against blood groups, tick transmissibility of vaccine strains, and the need for a cold chain transportation. Vaccinated cattle should be housed or kept under close observation for a month in case excessive reactions occur. A major problem in vaccination with live Babesia is the occasional apparent failure to transmit the protozoa. This may be result from the absence of the protozoa from the bloodstream of the donor at the time that the blood is drawn or from the presence of a prophylactic drug—for example, imidocarb dipropionate—or low levels of antibody in the animal’s tissues. Revaccination is necessary in these circumstances, preferably with blood from a donor that is undergoing a severe reaction at the time. The attenuated organisms used in unfrozen South Africa B. bovis and B. bigemina vaccines are susceptible for longer periods to the residual effect of the antibabesial drugs diminazene and imidocarb dipropionate compared with the virulent field strains. The waiting periods before administration of the frozen B. bovis and B. bigemina vaccines in animals that have been treated with diminazene at 3.5 mg/kg BW compare favorably with those of unfrozen vaccines at 4 and 8 weeks. The inhibitory effect of imidocarb dipropionate at 3.0 mg/kg BW on the infectivity of both frozen B. bovis and B. bigemina vaccines is longer and requires minimum waiting periods before administration of these vaccines of 12 weeks and 24 weeks, respectively. Vaccination With Subunit Vaccines Subunit vaccines offer an attractive alternative to virulent or attenuated Babesia spp.9,10 Such vaccines are based on recombinant antigens derived from cloned complementary DNA from protozoan parasites. Several protective antigens associated with merozoites or merozoite-infected erythrocytes of B. bigemina and B. bovis were identified as

possible molecules.10 Rhoptry-associated proteins might become targets of generic recombinant vaccines. Dead Vaccines Dead vaccines would overcome many of the inherent difficulties in the production, transport, and use of live vaccines.10 However, they have not been sufficiently efficacious, and more research is required. Vector Control This approach was first used successfully to control and eventually eradicate the cattle tick Boophilus annulatus and Babesia from the United States.1 In 1906, an eradication program began that involved livestock owners, state officials, and U.S. Department of Agriculture specialists. The program involved three tactics. First, some pastures were rendered tick-free by excluding all host animals until the ticks had starved to death. The second, more common tactic was to retain livestock on the infested pastures and to disinfect the animals at regular 2-week intervals by immersion in an arsenic solution, which killed the engorged female ticks. Third, the interstate movement of tickinfested cattle was prohibited through quarantine. The campaign to eradicate cattle ticks from the United States is the most sustained, extensive, coordinated area-wide attack ever made against an arthropod pest. The tick was removed from more than a million square kilometers over a period of 34 years. The tick is confined to the lower Rio Grande River in Texas, where reinfestation occurs via animal movement from Mexico. This necessitates continual control of fringe populations of cattle. In Africa, babesiosis is only part of very important complexes of ticks and tick-borne disease, and intensive government-regulated tick control programs have been used for many years. In other continents, the situation is much less complex; where babesiosis is endemic, disease control (rather than eradication) is more realistic. Eradication of tick vectors is rarely considered practical, environmentally sustainable, or economically justifiable on either a national or a local basis. Natural Endemic Stability Natural endemic stability can seldom be relied upon on as a disease control strategy.9 First, in endemic areas, climatic effects, genetic makeup of hosts, and management strategies inevitably have a major effect on the rate of transmission and, ultimately, on the likelihood of endemic stability developing. Second, endemic stability is an economic concept that incorporates risk management and loss thresholds. The climatic, animal, and management parameters that allow endemic stability can change on a seasonal and annual basis. Third, the model for endemic stability was developed in

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

Australia and the Americas, where disease/ vector interactions are relatively simple. The African situation is much more complex and less predictable, with four main diseases, several vectors, the presence of game reservoirs, and a larger range of susceptibility of bovine breeds. Control of Babesiosis in Species Other Than Cattle The principles of the control of babesiosis in other species is similar to those used for control of this disease in cattle. Most attention is focused on controlling the vector tick, identifying infected and carrier animals by an appropriate laboratory test, and sterilizing the positive reactors using a suitable treatment strategy. FURTHER READING Mueller J, Hemphill A. In vitro culture systems for the study of apicomplexan parasites in farm animals. Int J Parasitol. 2013;43:115-124. Suarez CE, Noh S. Emerging perspectives in the research of bovine babesiosis and anaplasmosis. Vet Parasitol. 2011;180:109-125.

REFERENCES

1. Radostits O, et al. Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1483. 2. Uilenberg G. Vet Parasitol. 2006;138:3. 3. Gohil S, et al. Int J Parasitol. 2013;43:125. 4. Brown WC, et al. Vet Parasitol. 2006;138:75. 5. Allred DR, Al-Khedery B. Vet Parasitol. 2006;138:50. 6. Hunfeld KP, et al. Int J Parasitol. 2008;38:1219. 7. Mosqueda J, et al. Curr Med Chem. 2012;19:1504. 8. Vial HJ, Gorenflot A. Vet Parasitol. 2006;138:147. 9. De Waal DT, Combrink MP. Vet Parasitol. 2006;138:88. 10. Brown WC, et al. Parasite Immunol. 2006;28:315.

EQUINE PIROPLASMOSIS Piroplasmosis of equids is a tick-borne infectious disease caused by the hemoprotozoan parasites Theileria equi and Babesia caballi.1–12 Piroplasmosis, also known as equine babesiosis (B. caballi), theileriosis (T. equi), or “biliary fever,” affects all equids, including horses, donkeys, mules, and zebras. Infection with either or both of these obligate intraerythrocytic organisms can cause varying degrees of hemolytic anemia and associated systemic disease. Recently T. equi infection has reemerged in the United States; consequently, questions have arisen as to the tick–vector– parasite–host relationship required for the development of clinical disease. SYNOPSIS Etiology Babesia caballi and Theileria equi. Epidemiology Occurs in equids. Transmission by ticks. Clinical signs Anemia, hemoglobinuria, jaundice, fever, often high case-fatality rate for T. equi but not for B. caballi.

Clinical pathology Parasites in stained blood smear, positive serology. Polymerase chain reaction (PCR) can be used for detection of parasite in blood or tissues. Necropsy lesions Thin, watery blood; pallor; jaundice. Diagnostic confirmation Parasites in blood smear; vector present in environment. Differential diagnosis list  A syndrome of acute hemolytic anemia should suggest the following alternative diagnoses: Equine infectious anemia Severe myoglobinuria (rhabdomyolysis associated with exercise or hypoglycin A intoxication) Foals with alloimmune hemolytic anemia Cardiac form of African horse sickness Treatment Imidocarb—drug of choice. (R1) Aspects of control Tick control; chemotherapy with imidocarb; surveillance of horses and ticks using effective serologic, molecular, and complementary tools.

ETIOLOGY B. caballi and T. equi (previously Babesia equi) are known to cause infections and disease in equids, including horses, donkeys, mules, and zebras.1–4 For T. equi, molecular studies support previous observations of preerythrocytic stages in lymphocytes. Nonetheless, the taxonomy of T. equi remains controversial.5

EPIDEMIOLOGY Geographic Occurrence The distribution of the causative protozoa is governed by the geographic and seasonal distribution of the insect vectors that transmit them (Table 11-10). Host Occurrence Babesiosis and theileriosis of equids are known as equine piroplasmosis. In horses, donkeys, mules, and zebras the disease is associated with B. caballi or T. equi.1–4 The diseases caused by B. caballi and T. equi are similar clinically, but the latter species is more virulent.3 Equine piroplasmosis occurs in much of southern Europe, Asia, and the Americas.3 For example, it is widespread in China and a cause for serious concern in northeastern China. In addition, equine piroplasmosis is also widespread in horses, mules, donkeys, and zebras in South Africa, and has reemerged in the United States.6–8 Australia is free from equine piroplasmosis, but seropositive horses were temporarily imported into Australia for the Sydney Olympic games in 2000. While in Australia, seropositive horses were kept at particular restricted sites. The distribution of equine piroplasmosis coincides with the distribution of tick vectors.2–4 Ixodid ticks of the genera Hyalomma, Dermacentor, and Rhipicephalus

811

have been identified as vectors for the transmission of either T. equi or B. caballi to equid hosts. In tropical countries, Hyalomma species appear to be suitable vectors for transmission of T. equi to horses and donkeys. Impact Mortality rates in outbreaks of equine piroplasmosis can be high, but the predominant losses in horses result from the interference with racing and equine competitions and meetings. This is a particular issue now, with the increased movement of horses between or among countries to partake in international equine competitions. Another possible form of loss is the death of foals infected in utero. Although there was early evidence that equine babesiosis might be an emerging disease threatening to be of major importance to the horse industry, this has not been the case. However, disease caused by T. equi appears to be a significant threat. Nonetheless, the use of diagnostic methods to screen horses before, during, and after international travel associated with international competitions or races is an important disease monitoring/prevention approach. Life Cycle and Transmission The life cycles of both T. equi and B. caballi involve distinct developmental stages that occur in the host and tick.3 Both parasites progress via three stages: the sporozoite (asexual transmission stage), merozoite (asexual blood stage), and gametocyte (sexual blood stage). Development within the tick varies, depending on the tick species involved. Infective sporozoites are transmitted through tick saliva to the equid host. Once within the equid host, B. caballi sporozoites directly invade erythrocytes, where they multiply and develop into trophozoites, and then into merozoites. After erythrocyte rupture, merozoites are released and invade other erythrocytes. In contrast, T. equi first enters peripheral mononuclear cells (PBMCs), which is part of the reason for its taxonomic reclassification as Theileria. Within PBMCs, T. equi zoites replicate to produce large schizonts; after ~ 9 days, merozoites are released and invade erythrocytes, where they multiply and develop into trophozoites, and then into merozoites. Merozoites are released and invade other erythrocytes. For both B. caballi and T. equi, asexual replication results in a massive expansion of the population of merozoites and parasitized erythrocytes. Following multiple rounds of replication, some merozoites develop into gametocytes within peripheral blood. Upon ingestion by a competent tick, the parasites undergo sexual reproduction, with gametocytes developing into gametes, which combine to form zygotes within the midgut of the tick. After 6 to 24 days, sporozoites accumulate within the salivary gland of the tick.

812

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Transmission can also occur iatrogenically through inappropriate mixing of the infected and uninfected blood.3 This can occur during the practice of sharing needles among different horses, but use of any bloodcontaminated equipment could result in transmission. Infection can also result when chronically infected horses serve as blood donors to naive horses. The illegal practice of blood doping (prerace blood transfusions) was implicated in an outbreak in Florida in 2008. Experimental infection can be induced by the IV and SC injection of infective stages and by infected ticks. Movement of Horses The international movement of animals has become a very important matter to the horse industry.3 These days, groups of pleasure horses are transported around the world to compete in different countries, and valuable stallions are sometimes transported to another country for a brief period to stand at stud. There is a tendency for some countries to be very restrictive in their quarantine procedures for horses; international relations might be enhanced if more was known about the relationship between a positive serologic test result and infection risk for other horses.

PATHOGENESIS

Although some aspects of pathogenesis remain unknown, infection with T. equi or B. caballi causes the lysis of erythrocytes, resulting in varying degrees of hemolytic anemia.3,4 The physical rupture of erythrocytes during the release of merozoites causes intravascular hemolytic anemia. Infected red blood cells are removed from the circulation by splenic macrophages, further contributing to hemolytic anemia. Uniformly, infection with T. equi results in more severe clinical disease than B. caballi.3 Nonparasitized erythrocytes are also removed from circulation, but the reason for this removal is unknown. It appears that the structure of erythrocyte membranes alters substantially during T. equi infection, suggesting that this change causes decreased deformability of the red cells, which might lead to a reduced microvascular blood flow. The level of malondialdehyde (marker of lipid peroxidation) in blood is significantly increased, suggesting that an accumulation of oxidative ions also contributes to erythrocyte lysis. T. equi and B. caballi infections also alter coagulation; B. caballi–infected erythrocytes cause microthrombi by clumping within small vessels, leading to venous stasis and vasculitis. Also described are thrombocytopenia and prolonged clotting times during T. equi and B. caballi infections. Decreased platelet counts might relate to immune-mediated destruction, splenic sequestration, and/or excess consumption, as seen in disseminated intravascular coagulation (DIC). Severe piroplasmosis often

results in hypercoagulability, systemic inflammatory response syndrome, and subsequent multiorgan system dysfunction. Placental transmission can result in abortion (usually in late gestation), stillbirth, or neonatal infection. Variation in genotypes of host and/or parasite could influence the prevalence of placental transmission. Transmission appears not to be linked to exposure to semen from an infected stallion, but blood contamination during mating might present a transmission risk. In most cases, horses become persistently infected and become carriers. The inapparent carrier state is life-long with T. equi and possibly for B. caballi. Persistent subclinical infection might result, in part, from the sequestration and immune-evasion strategies of the parasites. A carrier status represents a delicate balance between protozoa and immune responses; this balance is readily disturbed by the stress of transport, deprivation of food, pregnancy, or intercurrent disease. After transmission, depending on many factors, including infectious dose and immune status of the host, clinical signs usually develop within 12 to 19 days for T. equi and 10 to 30 days for B. caballi. The fatality rate of naive horses in geographic regions endemic for equine piroplasmosis appears to be 5% to 10%, but the severity of disease can vary significantly from one geographic region to another. Immunology The responses of the equine immune system to infection with T. equi or B. caballi are not yet completely understood but are undoubtedly complex and multifaceted and involve both cellular and humoral components.3,9,10 It is well accepted that infection with either parasite results in carrier status, which confers protection against disease. There is no documented cross-protection between T. equi and B. caballi, and horses can be infected with both parasites simultaneously.

CLINICAL FINDINGS

Clinical disease can present in different forms.3 For acute T. equi infection, clinical signs usually relate to hemolysis and resultant anemia. Although B. caballi–infected horses do become anemic, the rare cases of acute death from B. caballi appear to results from multiple-organ dysfunction linked to systemic formation of microthrombi and DIC. In these cases, clinical signs vary depending on the organ system affected. Horses with acute infection initially develop nonspecific signs, such as high fevers, sometimes greater than 40° C (104° F), lethargy, peripheral edema, anorexia, and/or weight loss. Petechia caused by thrombocytopenia can be observed on mucous membranes, including the nictitating membrane. Signs of hemolytic anemia follow and include icteric or pale mucous membranes,

tachycardia, tachypnea, pigmenturia (linked to bilirubinuria or hemoglobinuria), and weakness. Some horses show gastrointestinal signs, including colic or impactions, followed by diarrhea. Other less common clinical signs include a secondary development of pneumonia, pulmonary edema, cardiac arrhythmia, catarrhal enteritis, laminitis, and/or central nervous system signs, characterized by ataxia, myalgia, and/or seizures. Permanent or temporary infertility has been seen in stallions. Acute renal failure might occur as a result of hemoglobininduced pigment nephropathy. Systemic responses to severe inflammation (hypotension) can worsen the kidney disease. Severe infections can also culminate in liver failure or DIC. A fulminant, abrupt onset of signs of (peracute) disease has been described. Sudden death from T. equi can occur, and the introduction of naive horses into an endemic region can lead to a rapid onset of a severe disease outbreak. Neonatal foals infected in utero with T. equi might present with severe and acute signs. Such foals can exhibit clinical signs at birth or can become ill at 2 to 3 days of age. Clinical signs, such as decreased suckling and weakness, are often nonspecific, but progress to resemble those of an infected adult horse (icterus, fever, and anemia). Cases of fetal and neonatal B. caballi infection have been reported but are rare. Chronic T. equi or B. caballi infection usually results only in nonspecific signs, including weight loss, poor performance, partial anorexia, and/or lethargy. Mild anemia might be present, and the spleen might be enlarged. Splenomegaly appears to be caused by an increased rate of extravascular hemolysis within the spleen in less severely affected horses. Pregnancy in carrier mares can result in abortion or neonatal infection. Because inapparent carriers can serve as reservoirs for transmission (via ticks, placentally or iatrogenically), such horses represent the largest challenge in nonendemic areas.

CLINICAL PATHOLOGY

Acute infection is characterized by severe leukocytosis, lymphopenia, and a high absolute neutrophil count. T. equi is detected in neutrophils and monocytes in the case of high parasitemia, indicating phagocytosis of the infected erythrocytes. Animals that die of T. equi infection show varying degrees of emaciation, hepato- and splenomegaly, and “flabby” kidneys. Petechial hemorrhages are also common in the liver, in the spleen, and on the cortical surface of the kidneys. Microscopic Examination Light microscopic examination of Giemsastained blood smears can be used to identify piroplasms within erythrocytes.3 T. equi and

Enzootic Bovine Leukosis (Bovine Lymphosarcoma)

VetBooks.ir

B. caballi can be readily distinguished from one another. Within erythrocytes, B. caballi typically appears as two large pyriform (pear-shaped) merozoites that measure 2 to 5 µm in length; the percentage of infected erythrocytes is typically less than 1%. T. equi merozoites occur within erythrocytes as small polymorphic piroplasms, occasionally in a distinct Maltese cross formation; T. equi merozoites usually measure 2 to 3 µm in length. In diseased horses, the percentage of infected erythrocytes is usually 1% to 5%, but it can be greater than 20% in severe cases. Serologic and DNA-Based Methods Various diagnostic techniques can be used alone or in combination.3 During an outbreak in a nonendemic region, involvement of the state and national regulatory agencies is essential. Only a few laboratories in the world are authorized to carry out specific testing for equine piroplasmosis; proper handling and dispatch of samples are crucial. IFAT is used as an adjunct assay to compare CFT results, and it is one of the prescribed tests for equine piroplasmosis recommended by the OIE. Western blot (or immunoblot) has also been used for the diagnosis of T. equi and B. caballi infections in a research setting, but is now increasingly used as a tool for the detection of B. caballi infection. Research is under way to critically validate these serologic/immunologic tests for use in routine diagnosis. The cELISA is one of the regulatory tests prescribed by the OIE for international horse transport. This test is considered to be the most sensitive means of detection of chronic T. equi infection. A cELISA for T. equi utilizes a recombinant protein (EMA-1; immunodominant, highly conserved surface antigen specific for T. equi) and specific monoclonal antibodies. This test has high sensitivity and specificity compared with all other serologic tests assessed to date. For T. equi, both the sensitivity and specificity of cELISA are greater than 95%. A cELISA using a recombinant protein (RAP-1) was also developed for B. caballi. However, sequence heterogeneity in RAP-1 among parasite strains is linked to an inability of the test to detect infected horses in some regions of the world.3,10 Both cELISAs are available commercially, but they are not available to practitioners. ELISA using whole T. equi merozoite antigen has been assessed and appears to be as an easy, economical, and reliable test. Although PCR and cELISA show considerable promise as diagnostic tools,3,11 more studies are needed to ensure adequate diagnostic specificity and sensitivity for routine application in different countries. Nonetheless, PCR assay can detect T. equi and B. caballi in the blood of horses that have recovered from acute babesiosis. Nested PCR has been used for the detection of T. equi and B. caballi in the blood samples from horses and

infections in ticks. Provided reliable genetic markers are employed, it should be possible to develop a PCR for routine diagnostic testing.

NECROPSY FINDINGS

In acute cases of equine piroplasmosis, in which patients die after a brief illness and during an anemic crisis, the typical signs include jaundice; thin, watery blood; pale tissues; enlargement of the spleen, which has a soft, pulpy consistency; and gross enlargement and dark-brown discoloration of the liver. The gallbladder is distended, with thick, granular bile; the kidneys are enlarged and dark; and the bladder contains red– brown urine. Petechial or ecchymotic hemorrhages are present under the epicardium and endocardium, and the pericardial sac contains an increased quantity of bloodstained fluid. A characteristic manifestation is severe DIC. In subacute or chronic cases of relatively long duration, the carcass is emaciated but hemoglobinuria is absent; the other changes observed in acute cases are present but are less pronounced. The microscopic examination of blood smears taken from peripheral blood and from kidney and heart muscle, and from the brain in the case of suspected disease, is mandatory for clinching the diagnosis. Smears from blood and most tissues must be made within 8 hours of death, within 28 hours for the brain, and stained with Giemsa for the detection of piroplasms. Direct fluorescent antibody staining of smears permits the use of slightly “older” tissues. Organ smears are still usable 5 days after collection, provided that they are kept at 22° C (72° F). The morphology of piroplasms can change quickly after the host’s death. Blood collected after death can also be used for detection of serum antibodies in serologic tests or parasite DNA by PCR. DIFFERENTIAL DIAGNOSIS Preferably, the presence of the tick vector should be collected and verified before a definitive diagnosis of piroplasmosis can be made, unless an animal has left a known enzootic area within preceding months. Clinically, a high morbidity and case fatality-rate in cases displaying jaundice with hemoglobinuria and fever are suggestive, but confirmation of the diagnosis by microscopic examination of stained blood smears, complementary immunologic or molecular tools, and/or by transmission experiments is required. A necropsied animal with splenomegaly, jaundice, hemoglobinuria, swollen and dark kidneys and liver, and/or myocardial petechia/ecchymoses is suggestive of babesiosis/piroplasmosis, but the diagnosis needs to be confirmed by traditional or molecular laboratory testing of tissues for the presence of the parasite stages (piroplasms).

813

Differential diagnosis list A syndrome of acute hemolytic anemia should suggest the following alternative diagnoses (Table 11-1): • Equine infectious anemia—has a much longer, recurrent course; usually occurs in sporadic cases; and is not associated with protozoa in body fluids and tissues • Myoglobinuria—red urine is a result of myoglobinuria, always associated with elevation of serum creatine phosphokinase activity • Foals with alloimmune hemolytic anemia— detectable only upon laboratory examination for evidence of incompatibility between the serum of the dam and the foal’s erythrocytes • Other immune-mediated disorders • Cardiac form of African horse sickness (AHS)—edematous lesions that occur are similar to those of babesiosis, but there is no evidence of hemoglobinuria or jaundice • Equine viral arteritis virus, equine anaplasmosis, purpura hemorrhagica, and red maple leaf toxicity

TREATMENT Chemotherapy For the alleviation of clinical signs, several drugs have been used with success, yet imidocarb, in its diproprionate salt form, is considered to be the most effective.3,12 Imidocarb, a carbanilide derivative, is typically administered to horses intramuscularly. The alternate form of this drug, a dihydrochloride salt, causes more severe muscle damage at the site of injection. Reported dosages of imidocarb for the alleviation of clinical signs vary, but 2.2 to 4.4 mg/kg given IM once is effective. If necessary, lower dosages can be repeated at 24- to 72-hour intervals for two to three treatments. In nonendemic regions, where chemotherapeutic clearance is desired, T. equi and B. caballi should be treated with 4.4 mg/kg of imidocarb IM every 72 hours (four times). Donkeys and mules are very sensitive to imidocarb; therefore, its use in these species is not recommended. Imidocarb has anticholinesterase activity, such that reactions to the drug might present as sweating, signs of agitation, colic, and/or diarrhea. Typically, these signs are transient and rarely life-threatening. Effects can be prevented with an IV dose of glycopyrrolate at 0.0025 mg/kg once, or reversed with a single IV dose of atropine at 0.2 mg/kg. Horses undergoing treatment should be monitored for complications and transient elevations of liver enzyme activities (aspartate aminotransferase [AST], alanine aminotransferase [ALT], alkaline phosphatase [ALP], and sorbitol dehydrogenase [SDH])—usually, these issues resolve following imidocarb treatment. Diminazene aceturate and diminazene diaceturate have also been used against T. equi and B. caballi at a dose of 3.5 mg/kg IM every 48 hours (two treatments).

814

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

Diminazene aceturate is more effective than diminazene diaceturate; both drugs have been reported to cause damage at the injection site. Efficacy of both drugs increases with the second dosage; no chemosterilization has been reported. Signs of toxicity include respiratory distress and lethargy. The antibiotic oxytetracycline when administered IV at a dose of 5 to 6 mg/kg, once daily for 7 days, is effective against T. equi but not against B. caballi. Other drugs reported to have efficacy in the treatment of babesiosis include amicarbilade isethionate, euflavine, artesunate and artemether (artemisinin derivatives), buparvaquone, and atovaquone, but these drugs are no longer used in practice.

5. Kappmeyer LS, et al. BMC Genomics. 2012;13:603. 6. Short MA, et al. J Am Vet Med Assoc. 2012;240:588. 7. Ueti MW, et al. PLoS ONE. 2012;7(9):e44713. 8. Scoles GA, et al. Emerg Infect Dis. 2011;17:1903. 9. Ramsay JD, et al. PLoS ONE. 2013;8(10):e76996. 10. Awinda PO, et al. Clin Vaccine Immunol. 2013;20:1752. 11. Baptista C, et al. Ticks Tick Borne Dis. 2013;4:242. 12. Grause JF, et al. Vet J. 2013;196:541.

Supportive Treatment In addition to the use of antiprotozoal drugs, acutely infected horses often require supportive treatment, including, but not limited to, intravenous fluids, NSAIDs, pain management, and blood transfusions. Adequate hydration is essential during treatment with imidocarb.3

Epidemiology Young animals on milk diet; most commonly nursing piglets that have not received supplemental iron. Housed nursing lambs. Occurs in veal calves fed milk with limited quantities of iron. Continued blood loss as a result of hemorrhage (lice, blood-sucking helminths). Subclinical iron deficiency occurs in calves and foals but is of doubtful significance. May be more susceptible to infectious diseases.

CONTROL

The principles of the control of piroplasmosis are similar to those used for the control of babesiosis in cattle. Most attention is focused on controlling the vector tick, identifying infected and carrier animals by an appropriate laboratory test, and the use of a suitable treatment strategy for test-positive animals. The control of ticks in pleasure horses by periodic spraying/treatment and inspection is a practical proposition when the animals are in constant use. No vaccines have been produced for use in horses. Chemotherapeutics that aid in the control of acute parasitemia and associated clinical signs, but do not eliminate infection, are important in endemic regions.3 In nonendemic regions, the goal is to maintain an infection-free status; therefore, when infected horses are detected, a safe chemotherapeutic with high efficacy for eliminating persistent infection needs to be administered. Accurate diagnostic tools and detailed knowledge of tick populations and their ability to transmit B. caballi and T. equi are essential for prevention of outbreaks in nonendemic regions.3 Increasing globalization of the equine industry and a changing climate provide challenges for the prevention and control of T. equi and B. caballi. Disease surveillance and detailed knowledge of vector competence and habitat through the use of effective molecular tools will be essential. REFERENCES

1. Radostits O, et al. Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1483. 2. Uilenberg G. Vet Parasitol. 2006;138:3. 3. Wise LN, et al. J Vet Intern Med. 2013;doi:10.1111/ jvim.12168; [Epub ahead of print]. 4. Kumar S, et al. Jpn J Vet Res. 2009;56:171.

Nutritional Deficiencies IRON DEFICIENCY SYNOPSIS Etiology Dietary deficiency of iron.

Signs Pale white skin of well grown nursing piglets, dyspnea, pallor of mucosae, sudden death may occur. Stillbirths if sows iron deficient. Secondary infectious diseases. Clinical pathology Subnormal levels of hemoglobin and serum iron; microcytic hypochromic anemia. Necropsy findings Pallor; thin. watery blood; anasarca; dilated heart; enlarged liver. Diagnostic confirmation Low serum hemoglobin and serum iron with microcytic hypochromic anemia. Response to iron therapy. Differential diagnosis Other causes of anemia (Table 11-3). Treatment Parenteral and oral iron salts. Control Ensure adequate iron intake. Parenteral iron-dextran to nursing piglets and lambs.

ETIOLOGY Iron deficiency is usually primary and most likely to occur in newborn animals whose sole source of iron is the milk of the dam because milk is a poor source of iron. Deposits of iron in the liver of the newborn are insufficient to maintain normal hemopoiesis for more than 2 to 3 weeks and are particularly low in piglets.

EPIDEMIOLOGY

Iron-deficiency states are not common in farm animals except in the very young confined to a milk diet. Iron-deficiency anemia occurs in nursing piglets for three reasons: 1. They do not have access to soil, which is a main source of iron for young farm animals.

2. They grow rapidly, and thus their absolute requirements for iron are high. 3. Milk is a poor source of iron. The administration of iron dextran to the piglets at a few days of age is preventive and is a routine health management strategy in modern pig production. If they do not receive supplemental iron dextran, clinical disease occurs usually when the piglets are 3 to 6 weeks old. The losses that occur include those resulting from mortality, which may be high in untreated pigs, and to failure to thrive. Under modern pig production systems, piglets do not have access to sufficient dietary iron until they are weaned to a dry diet containing supplemental iron, thus the need for administration of parenteral iron dextran to all piglets at a few days of age. Even piglets raised outdoors with access to soil perform better when supplemented with iron. Iron-injected piglets raised outdoors are heavier at weaning, there is less preweaning morbidity and mortality, and they have higher blood hemoglobin concentrations compared with nonsupplemented piglets.1 Larger piglets in a litter appear to be at greater risk of developing iron deficiency at weaning.2 Iron deficiency in pigs increases the severity of Trichuris suis and Ascaris suum infections. Iron-deficiency anemia occurs in nursing lambs that are housed and do not have access to soil, do not consume much feed other than their dam’s milk for the first 7 to 10 days of life, and grow at 0.4 kg/d. The parenteral administration of iron dextran at 24 h of age prevents the anemia. Abomasal bloat occurs in these lambs with lower serum iron concentration, and iron-dextran injections are preventive and also have a significant effect on weight gain and red blood cell and iron parameters. Continued blood loss by hemorrhage in any animal may result in subclinical anemia and iron deficiency. Cattle heavily infested with sucking lice may develop serious and even fatal anemia. The chronic form is characterized by a nonregenerative anemia with subnormal levels of serum iron, and treatment with iron is necessary for an optimal response. Horses carrying heavy burdens of bloodsucking strongylid worms often have subnormal hemoglobin levels and respond to treatment with iron. Occasionally veal calves, and possibly young lambs and kids, may also suffer from an iron deficiency. Good-quality veal is traditionally pale in color and is produced by feeding calves an all-liquid milk replacer diet with a low concentration of available iron. The pallor of veal is largely a result of low concentrations of myoglobin and other iron-containing compounds in muscle. use of milk replacers containing only 10 mg iron/kg DM results in marked anemia and reduced growth performance. Feeding milk replacers with 50 mg iron/kg DM is considered, physiologically,

Nutritional Deficiencies

VetBooks.ir

the optimum amount of iron for veal calves but may be too high for acceptable carcass yield in some countries. A severe iron deficiency with reduced growth rate in veal calves may be associated with a higher incidence of infectious disease because of an impaired immune system. The objective in veal calf management is to walk the narrow line between the maximum production of white meat and a degree of anemia insufficient to interfere with maximum production. Subclinical iron-deficiency anemia also occurs in newborn calves and kids, but there is debate as to whether the condition has practical significance. Supplementation of dairy calves with iron, or iron and copper, increases growth rate.3 In newborn calves affected with a normochromic, normocytic, and poikilocytic anemia, the levels of serum iron are not significantly different from those of normal calves. It has been proposed that severe poikilocytosis in calves is associated with abnormalities of hemoglobin composition and protein 4.2 in the erythrocyte membrane, and iron deficiency is the cause of moderate poikilocytosis in calves. Anemia, without clinical signs, is most likely to occur when calves are born with low hemoglobin and hematocrit levels, a relatively common occurrence in twins. It is possible that suboptimal growth may occur during the period of physiologic anemia in early postnatal life. There is some evidence for this in calves in which hemoglobin levels of 11 g/dL at birth fall to about 8 g/dL between the 30th and 70th days and only begin to rise when the calves start to eat roughage. The daily intake of iron from milk is 2 to 4 mg in calves, and their daily requirement during the first 4 months of life is of the order of 50 mg, so that iron supplementation of the diet is advisable if the calves are fed entirely on milk. Even when hay and grain are fed to calves and lambs in addition to milk, there is a marked growth response to the administration of iron-dextran preparations at the rate of 5.5 mg/kg BW. The dietary iron requirement for fast-growing lambs is between 40 and 70 mg/kg BW, and growth rate is suboptimal on diets of less than 25 mg/kg BW. Low serum iron concentration and low serum ferritin have been observed in hospitalized young foals. Microcytic anemia and hypoferremia occur in Standardbred foals kept at pasture for 12 hours per day. These changes are not prevented by oral administration of four oral doses of 248 mg of iron, suggesting that higher levels of supplementation are needed. Conversely, hypoferremia and anemia were reported in stabled foals but not in a pastured cohort. The stabled foals had clinical signs of anemia (lethargy) and low hematocrit, hemoglobin, and serum iron concentrations, which were restored to normal values by iron supplementation (0.5 g iron sulfate orally once

815

daily, 3 g of iron sulfate top dressed on cut pasture fed to the foals and their dams, and unlimited access to a lick block containing iron). Although the colostrum of mares is rich in iron, milk has much lower concentrations, probably explaining the low serum iron of some nursed foals and demonstrating the need for access to iron supplements or, preferably, soil or pasture. Supplementation of foals with iron should be undertaken cautiously because of the documented hepatotoxicity of large doses of iron given orally to newborn foals. Toxic hepatopathy develops in newborn foals administered iron fumarate at 16 mg/kg BW within 24 hours of birth, similar to the situation in piglets. Competition horses are frequently given iron supplementation to treat anemia and to improve performance, despite the fact that neither application has any scientific basis. In contrast, iron overload and toxicity have occurred in competition horses. Some studies have shown high total plasma iron in British 3-day event team horses before transport (77 µmol/L compared with normal levels of 24 µmol/L). Immediately after traveling for 3 days on the road, the plasma levels had declined to 29 µmol/L. The iron-binding antioxidant activity, an indicator of transferrin saturation, had also declined, suggesting greater saturation of available transferrin in the plasma or a decreased capacity to sequester iron. The saturation of mechanisms to sequester iron, such as may occur with excessive supplementation, may predispose the horses to iron-catalyzed oxidant injury. The total iron intake exceeded the normal recommendation of between 550 and 600 mg/d.4 Anemia (or a low packed cell volume) is not synonymous with iron deficiency but is frequently associated with disease processes.5 In addition, iron deficiency is unlikely to occur in healthy horses. Calcium carbonate added to the diet of weaned and finishing pigs may cause a conditioned iron deficiency and a moderate anemia, but this effect is not apparent in mature pigs. Manganese may exert a similar antagonistic effect.

prevent anemia in them. Piglets with access to iron show a gradual return to normal hemoglobin levels starting at about the 10th day of life, but in pigs denied this access, the hemoglobin levels continue to fall. One of the important factors in the high incidence of anemia in piglets is the rapidity with which they grow in early postnatal life. Piglets normally reach 4 to 5 times their birth weight at the end of 3 weeks, and 15 times their birth weight at the end of 8 weeks. The daily requirement of iron during the first few weeks of life is of the order of 15 mg. The average intake in the milk from the sow is about 1 mg/d, and the concentration in sow’s milk cannot be elevated by feeding additional iron during pregnancy or lactation. Apart from the specific effect on hemoglobin levels, iron-deficient piglets consume less creep feed, and after the first 3 weeks of life they make considerably slower weight gains than supplemented piglets. Although specific pathogen-free pigs show a less marked response to the administration of iron than pigs reared in the normal manner, it is obvious that they need supplementary iron to prevent the development of anemia. Irondeficient piglets appear to be more susceptible to diarrhea at about 2 weeks of age than are piglets that have received iron. A marked impairment of gastric secretion of acid and chloride and atrophic gastritis occurs in iron-deprived piglets. Villous atrophy of the small intestine and changes in the gastrointestinal flora also occur in iron-deficient piglets, which may contribute to the increased susceptibility to diarrhea. In iron-deficient piglets, lymphocyte activity is impaired, resulting in a decrease in circulating B-lymphocyte numbers and decreased immunocompetence. Severe iron deficiency in veal calves is characterized by impaired growth and reduced feed intake and utilization. The growth rate is reduced only when hemoglobin concentrations fall below 70 g/L. The reduced growth rate may be a result of reduction in the half-life of growth hormone.

PATHOGENESIS

The highest incidence of iron-deficiency anemia in piglets occurs at about 3 weeks of age, but it can occur up to 10 weeks of age. Affected pigs may be well grown and in good condition, but the growth rate of anemic pigs is significantly lower than that of normal pigs, and feed intake is reduced. A mild diarrhea may occur, but the feces are usually normal in color. Dyspnea, lethargy, and a marked increase in amplitude of the apex beat of the heart can be felt after exercise. The skin and mucosae are pale and may appear yellow in white pigs. Edema of the head and forequarters, giving the animal a fat, puffed-up appearance, may be present. A lean, white, hairy look is probably more common. Death usually occurs suddenly, or affected animals may survive in a thin,

More than half the iron in the animal body is found as a constituent of hemoglobin. A relatively small amount is found in myoglobin and in certain enzymes that play a part in oxygen utilization. Piglets at birth have hemoglobin levels of about 90 to 110 g/L. A physiologic fall to 40 to 50 g/dL occurs in all pigs, with the lowest levels occurring at about the 8th to 10th day of life. Levels of iron in the liver at birth are unusually low in this species and cannot be increased appreciably by supplementary feeding of the sow during pregnancy. The IM injection of iron-dextran preparations to sows during late pregnancy does elevate the hemoglobin levels of the piglets during the first few weeks of life but not sufficiently to

CLINICAL FINDINGS

816

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

unthrifty condition. A high incidence of infectious diseases, especially enteric infection with E. coli, is associated with the anemia, and streptococcal pericarditis is a well-recognized complication. Under experimental conditions, similar signs occur in calves, and there is, in addition, an apparent atrophy of the lingual papillae. A high incidence of stillbirths is recorded in the litters of sows suffering from iron-deficiency anemia.

CLINICAL PATHOLOGY

The characteristic abnormality in iron-deficiency anemia is presence of hypochromic, microcytic red cells, although early in disease there can be macrocytic anemia as a result of chronic hemorrhage (Table 11-3). There is also reduced plasma or serum concentrations of ferritin and serum iron, increased total iron-binding capacity, and reduction in stainable iron in bone marrow. Anemia of iron deficiency must be differentiated from anemia resulting from chronic inflammatory disease (Table 11-3).5 In normal piglets there is a postnatal fall of hemoglobin levels to about 8 g/L and sometimes to as low as 4 to 5 g/L during the first 10 days of life. In iron-deficient pigs there is a secondary fall to 20 to 40 g/L during the third week. The hemoglobin level at which clinical signs appear in pigs is about 40 g/L. Erythrocyte counts also fall from a normal of 5 to 8 × 10 /L down to 3 to 4 × 10 /L and may be a better index of iron status than hemoglobin levels. Iron-deficiency anemia in piglets is a microcytic hypochromic anemia. In chronic blood-loss anemia in cattle infested with sucking lice, there is a nonregenerative anemia and a decrease in serum iron levels. Serum levels of iron considered to be normal in sheep and cattle are 100 to 200 µg/dL (17.9 to 35.8 µmol/L). In newborn calves, the levels are 170 µg/dL (30.4 µmol/L) at birth and 67 µg/dL (12.0 µmol/L) at 50 days of age. Serum ferritin concentration is an index for monitoring prelatent iron deficiency of calves. The borderline of iron-deficiency anemia of veal calves at 16 to 20 weeks of age has been defined as a hemoglobin concentration of 9 g/L and a saturation of total iron binding capacity of 10%.

NECROPSY FINDINGS

The carcass is characterized by pallor, watery blood, and moderate anasarca. The heart is always dilated, sometimes extremely so. The cardiac dimensions in severely anemic neonatal pigs indicate that dilatation and hypertrophy occur consistently. The liver in all cases is enlarged and has a mottled tan– yellow appearance. Histologic examination of the bone marrow reveals maturation asynchrony of the erythroid line and a lack of hemosiderin stores. Other microscopic changes described include periacinar hepatocellular changes typical of hypoxia and

decreased numbers of parietal cells in the gastric mucosa. Samples for Confirmation of Diagnosis • Toxicology—50 g liver (ASSAY [Fe]) (Note that serum ferritin from surviving littermates is a better indicator of iron status.) • Histology—liver, heart, bone marrow, stomach (LM) DIFFERENTIAL DIAGNOSIS Confirmation of the diagnosis will depend on hemoglobin determinations and curative and preventive trials with administered iron. The possibility that anemia in piglets may be caused by copper deficiency should not be overlooked, especially if the response to administered iron is poor. Isoimmunization hemolytic anemia can be differentiated by the presence of jaundice and hemoglobinuria, and the disease occurs in much younger pigs. Eperythrozoonosis occurs in pigs of all ages, and the protozoan parasites can be detected in the erythrocytes.

TREATMENT Principles of treatment are removal of the cause of iron loss (chronic bleeding, parasitism, inadequate diet) and provision of supplemental iron. The emphasis in treatment should be on removal of the inciting cause of iron deficiency. Supplemental iron should be provided to correct the whole-body deficiency of iron and can be achieved by oral or parenteral administration. Oral administration is preferred because it is safer, is less expensive, and does not require the expertise needed for intravenous or intramuscular administration. Parenteral administration of iron preparations is associated with severe tissue reactions and, with intravenous administration, acute death.

CONTROL

Preventive measures must be directed at the neonatal piglets because treatment of the sows before or after farrowing is generally ineffective, although some results are obtained if the iron preparations are fed at least 2 weeks before farrowing. Ferric choline citrate appears to have some special merit in this field. Allowing the nursing piglets access to pasture or dirt yards or periodically placing sods in indoor pens can offer adequate protection. Where indoor housing on impervious floors is necessary, iron should be provided at the rate of 15 mg/d until weaning, either by oral dosing with iron salts of a commercial grade or by the IM injection of organic iron preparations. These methods are satisfactory, but the results are not usually as good as when piglets are raised outdoors. However, indoor housing is practiced in many areas to avoid exposure to parasitic

infestation and some bacterial diseases, especially erysipelas. If sods are put into pens, care must be taken to ensure that these diseases are not introduced. Dietary Supplementation Sows Feeding sows a diet supplemented with 2000 mg iron/kg DM of diet will satisfactorily prevent iron-deficiency anemia in the piglets. The piglets will ingest about 20 g of sow feces per day, which will contain sufficient iron and obviate the need for IM irondextran injections. The piglets grow and thrive as well as those receiving the irondextran injections. Veal Calves Milk replacers for veal calves may contain up to 40 mg/kg DM of iron for the first months, but commonly contain only 10 to 15 mg/kg DM for the finishing period. The best indicator of the onset of anemia in calves on vealer diets is loss of appetite, which is a more sensitive indicator than biochemical measurement. Heifer Calf Herd Replacements The National Research Council recommends that milk replacers fed to herd replacements or dairy beef contain 100 mg/kg of DM, with an upper limit of 1000 mg/kg DM. The preruminant calf can tolerate between 2000 and 5000 ppm DM iron in milk replacer. Oral Dosing Daily dosing with 4 mL of 1.8% solution of ferrous sulfate is adequate. Iron pyrophosphate may also be used (300 mg/d for 7 days). To overcome the necessity for daily dosing, several other methods of administering iron have been recommended. A single oral treatment with iron-dextran or irongalactan has been recommended, provided that an excellent creep feed is available, but the method seems unnecessarily expensive. With this oral treatment it is essential that the iron be given within 12 hours of birth because absorption has to occur through the perforate neonatal intestinal mucosa; later administration is not followed by absorption. Reduced iron (British Veterinary Codex) can be administered in large doses because it does not cause irritation of the alimentary mucosa. A single dose of 0.5 to 1 g once weekly is sufficient to prevent anemia. Alternatively, the painting of a solution of ferrous sulfate on the sow’s udder has been recommended (450 g ferrous sulfate, 75 g copper sulfate, 450 g sugar, 2 L water—applied daily) but has the disadvantage of being sticky and of accumulating litter. Pigs raised on steel gratings can derive enough iron from them to avoid the need for other supplementation. Excessive oral dosing with soluble iron salts may cause enteritis, diarrhea, and some deaths in pigs. High intakes of ferric hydroxide cause diarrhea, loss of

Nutritional Deficiencies

VetBooks.ir

weight, and low milk production in cattle. The presence of diarrhea in a herd prevents absorption of orally administered iron, and treatment by injection is recommended in this circumstance. Intramuscular Injection of Iron Preparations Suitable preparations must be used and are usually injected IM in piglets on one occasion only, between the day 3 and day 7 of life. Iron-dextran, fumarate, and glutamate are most commonly used. A dose of 200 mg of a rapidly absorbed and readily utilizable form of iron within the first few days of life will result in greater body weights at 4 weeks of age than in piglets given only 100 mg. Multiple injections give better hemoglobin levels but have not been shown to improve weight gain, and thus a second injection at 2 to 3 weeks of age may not be economical. A total dose of 200 mg is usually recommended as being required to avoid clinically manifest iron-deficiency anemia, but to avoid any chance of a subclinical deficiency the feed should contain additional iron at the level of 240 mg/kg. A new preparation (Heptomer) contains 200 mg/mL of iron, permitting a full dose in one injection. Contrasting information is that one injection of 100 mg of iron is adequate for baby pigs. Acute poisoning and rapid death can occur in piglets given iron-dextran compounds parenterally if the piglets were born from sows that were deficient in vitamin E and selenium during gestation. This is discussed in the section on iron-dextran poisoning. In normal piglets, the iron-dextran compounds are safe and are usually not toxic even on repeated injection. These preparations are ideal for treatment because of the rapid response they elicit and the absence of permanent discoloration of tissues after their use if given during the first month of life. A combination of sodium selenite and iron-dextran has been given to piglets at 3 days of age and is superior to treatment with iron alone when the piglets are deficient in selenium. Iron supplementation should also be administered to suckling piglets raised outdoors. Iron-deficiency anemia in housed lambs is preventable by the IM injection of 300 mg iron dextran at 24 hours of age. At 12 and 24 days after treatment, the hematological values in the treated group were significantly different from those of the unsupplemented group, and at weaning, the treated lambs were 1.0 kg heavier than untreated lambs. An oral iron supplement given to these housed lambs improved red cell and iron parameters but did not improve performance. Comparable doses of parenteral irondextran compounds have been used for the treatment of iron-deficiency or iron-loss anemias in other species, but accurate doses have not been established, and the use of

these preparations in cattle and horses is expensive. In addition, iron-dextran preparations given IM to horses may cause death within a few minutes after administration. The most inexpensive method of supplying iron is to use ferrous sulfate orally at a dose of 2 to 4 g daily for 2 weeks to adult cattle and horses with iron-deficiency anemia. Iron injection of beef calves in the first week after birth will result in an increase in packed cell volume (PVC), hemoglobin (Hb), mean corpuscular volume (MCV), and mean corpuscular hemoglobin (MCH), which persists for 12 weeks. However, weight gains during the first 18 weeks of life were not affected. REFERENCES

1. Pearson R. Pig Journal. 2011;64:6. 2. Bhattarai S, et al. J Swine Health Prod. 2015;23:10. 3. Bami MH, et al. Vet Res Comm. 2008;32:553. 4. Nutrition NRCSoH. Nutrient requirements of horses: National Academies. 2007. 5. Borges AS, et al. J Vet Int Med. 2007;21:489.

COBALT DEFICIENCY Cobalt deficiency is a disease of ruminants ingesting a diet deficient in cobalt, which is required for the synthesis of vitamin B12 (cyanocobalamin) by rumen microflora. The disease is characterized clinically by inappetence and loss of body weight. Some effects on reproductive performance in sheep have been reported. Cobalt was first shown to be an essential nutrient for sheep and cattle following investigations in the 1930s of two naturally occurring diseases occurring on soils of Aeolian origin in Australia, coast disease of sheep and wasting disease of cattle.1 Soon after it was recognized that similar wasting disorders in several countries responded to cobalt supplementation, including cobalt pine in Scotland, salt sickness in Florida, Nakururitis in Kenya, and bush sickness in New Zealand.1 SYNOPSIS Etiology Dietary deficiency of cobalt resulting in a deficiency of vitamin B12. Epidemiology Occurs worldwide, primarily in cattle and sheep, where soils are deficient in cobalt and no supplements are given. Associated with ovine white liver disease and phalaris staggers. Signs Inappetence, gradual loss of body weight, pica, marked pallor of the mucous membranes, and lacrimation. Decreased wool growth, milk production, and lambing percentage. Clinical pathology Liver cobalt or vitamin B12 concentration, serum vitamin B12 (sheep). Elevated methylmalonic acid in plasma and urine; elevated formiminoglutamic acid in urine. Normocytic normochromic anemia. Necropsy findings Emaciation, hemosiderosis of spleen.

817

Diagnostic confirmation Location of farm and soil type. Vitamin B12 and cobalt of liver. Response to supplementation. Differential diagnosis list Common causes of ill-thrift in ruminants: General nutritional deficiency (protein and energy) Intestinal helminthiasis Copper deficiency Johne’s disease Treatment Oral dosing with cobalt or parenteral injections of vitamin B12. Control Dietary supplementation with cobalt (cobalt pellets, top dressing of pastures or foliar sprays). Long-acting preparations of vitamin B12.

ETIOLOGY The disease is caused by a deficiency of cobalt in the diet, which results in a deficiency of vitamin B12. Vitamin B12 is central to many metabolic pathways, including the conversion of propionic acid to glucose (the only direct source of glucose for ruminants) and the metabolism of methionine, which is essential for wool growth and the transport of folic acid into liver cells.

EPIDEMIOLOGY Occurrence Cobalt deficiency occurs in Australia, New Zealand, the United Kingdom, North America, the Netherlands, and probably in many other parts of the world. Where the deficiency is extreme, large tracts of land are unsuitable for the raising of ruminants, and in certain areas, suboptimal growth and production may be limiting factors in the husbandry of sheep and cattle. Historically, ill-thrift from cobalt deficiency was so marked that many calves and lambs died. In those that survived, growth rates were markedly depressed compared with cobalt-supplemented lambs. In most known deficient areas, signs of cobalt deficiency are now confined mainly in lambs because supplementation or cobalt fertilizer has been applied for many decades. However, cobalt-responsive ill-thrift in lambs still occurs where cobalt fertilizer applications or vitamin B12 injections to lambs are haphazard, with live weight gains following vitamin B12 injections of up to 180 g/d. The soils on which cobalt deficiency occurs are usually well drained but can be of quite diverse geological origin, and the concentration of cobalt in the soil can vary widely.1 Where cobalt deficiency occurs, soil cobalt is usually less than 2 mg/kg DM, and the available (extractable) cobalt is less than 0.25 mg/kg DM. The availability of cobalt to plants is reduced by high concentrations of manganese in the soil and heavy liming. Sheep are more susceptible to cobalt deficiency than cattle, and young animals are more susceptible than adults. The disease

818

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

occurs most commonly in ruminants grazing pasture in severely deficient areas, but sporadic cases occur in marginal areas. In Australia this can occur after certain seasonal conditions, such as the intensive application of fertilizer and high spring rainfall that produces a flush of pasture growth, or in Europe after long periods of stable feeding. In the latter scenario, bulls, rams, and calves are most commonly affected. A disease of moose called “moose sickness” occurs in eastern North America, mainly in the Tobeatic and Cape Breton Highlands of Nova Scotia in Canada. Low concentrations of cobalt and vitamin B12 are found in the liver and increased concentrations of methylmalonic acid in the plasma. There are striking similarities between the North American moose sickness and a moose disease in Sweden caused by molybdenosis. Cobalt deficiency is unlikely to occur in pigs and other omnivores or carnivores because vitamin B12 is present in meat and other animal tissues. Horses appear to be unaffected. Cobalt is also protective against the liver damage in sheep exposed to annual ryegrass and the neurologic signs induced in phalaris staggers. Risk Factors Dietary and Environmental Factors Pastures containing less than 0.07 and 0.04 mg/kg DM result in clinical disease in sheep and cattle, respectively. The daily requirement for sheep at pasture is 0.08 mg/ kg DM of cobalt, but growing lambs have a greater requirement (11 µg/d), and reduced growth is likely at pasture levels less than 0.10 mg/kg DM. Variations in the cobalt content of pasture occur with seasonal variations in pasture growth, and an increased incidence of deficiency in spring may be related to domination of the pasture by rapidly growing grasses, which have lower cobalt content than legumes. There is also a great deal of variation between years. Forage grown on well-drained soils has greater cobalt content than that grown on poorly drained soils of the same cobalt status. Although plant growth is not visibly affected by low soil cobalt, the addition of excessive quantities may retard growth. Primary cobalt deficiency occurs only on soils that are deficient in cobalt. Such soils do not appear to have any geological similarity, varying from windblown shell sands to soils derived from pumice, ironstone, and granite, and Japanese soils composed largely of volcanic ash are seriously deficient. The soils in New Brunswick, Canada, are naturally acidic, and the cobalt content of the soil is decreased by leaching associated with an annual rainfall of 120 cm. Surveys in this area show average values of 0.028 and 0.088 mg/kg DM for grasses and legumes,

respectively, which justifies supplementation of ruminant diets with cobalt. After the introduction of grazing livestock, large parts of New Zealand were found to be trace-element deficient (cobalt, selenium, and copper). Livestock grazing pastures grown on these soils may be deficient in one or more of these trace elements, and correcting these deficiencies is now a common and essential animal health management strategy. In New Zealand, soil types are categorized as severe, moderate, or marginal. Of the land considered suitable for farming, about 1 million hectares on the North Island and 918,000 hectares on the South Island are defined as cobalt deficient. Outbreaks of cobalt deficiency have occurred in cattle grazing on pastures on the granite-derived northern tablelands of New South Wales in Australia and in sheep grazing pasture on soils derived from weathered rhyolite and ignimbrite, the former being inherently low in cobalt. Cobalt deficiency still occurs in areas where it has previously never been diagnosed, and so in seasons of lush spring and summer pasture growth it should be included as a differential diagnosis when investigating cases of unthriftiness. In the northern Netherlands, lambs grazing cobalt-deficient pastures and not supplemented with cobalt are 6.7 times more likely to die than supplemented lambs. This occurs despite affected farms often having acetic acid–extractable soil cobalt content of pastures greater than the reference value for cobalt deficiency (≤0.30 mg Co/kg dried soil). Although soils containing less than 0.25 mg/kg cobalt are likely to produce pastures containing insufficient cobalt, the relationship between levels of cobalt in soil and pasture is not consistent. The factors governing this relationship have not been determined, although heavy liming and high concentrations of manganese reduce the availability of cobalt in the soil. Ovine White Liver Disease A specific hepatic dysfunction of sheep has been described in New Zealand, Australia, the United Kingdom, Norway, and in grazing lambs in the Netherlands. It is called white liver disease because of the grayish color of the liver. Clinically, it is manifested by photosensitization when the disease is acute and anemia and emaciation when the disease is chronic. It seems likely that the disease is a toxic hepatopathy (fatty liver degeneration) caused by the accumulation of methylmalonic acid. This is converted into branchedchain fatty acids, causing liver failure, hepatic encephalopathy, and photosensitization. It is prevented by adequate dietary cobalt. Hepatic Lipidosis in Goats Hepatic lipidosis of goats in Oman is associated with low serum vitamin B12 and low

liver cobalt and can be experimentally reproduced by a low intake of cobalt. It is one of the most frequent causes of liver condemnation and a significant economic loss because goats are the predominant domesticated animal reared for meat in Oman. Experimental Reproduction of Cobalt Deficiency in Sheep Cobalt deficiency can be reproduced in sheep by feeding diets containing less than 70 µg/kg cobalt. A diet containing 4.5 µg/kg fed to lambs produced a condition similar to naturally occurring cobalt deficiency, with subnormal plasma and liver concentrations of vitamin B12, reduced growth rate, serous ocular discharge, alopecia, emaciation, and fatty degeneration of the liver, which had reduced concentrations of vitamin B12 (14.5 pmol/g) at necropsy. The liver lesions included accumulation of lipid droplets and lipofuscin particles in hepatocytes, dissociation and necrosis of hepatocytes, and sparse infiltration by neutrophils, macrophages, and lymphocytes. Ultrastructural changes in hepatocytes included swelling, condensation and proliferation of mitochondria, hypertrophy of smooth endoplasmic reticulum, vesiculation and loss of arrays of rough endoplasmic reticulum, and accumulation of lipid droplets and lipofuscin granules in cytoplasm. Reduced activities of vitamin B12–dependent enzymes, methylmalonyl CoA mutase, and methionine synthesis, along with lipid peroxidation, are the likely mechanisms in the development of the liver lesions.

PATHOGENESIS

Cobalt is a unique essential trace element in ruminant nutrition because it is stored in the body in only limited amounts and not in all tissues. In the adult ruminants the only known function of cobalt is in the rumen, where participates in the production of vitamin B12 (cyanocobalamin), and it has to be continuously ingested in the feed. Ruminants have a much higher requirement for vitamin B12 than other species. In sheep, this is around 11 µg/d, with up to 500 µg/d produced in the rumen but most being lost. Animals in the advanced stages of cobalt deficiency are cured by the oral administration of cobalt or by the parenteral administration of vitamin B12. On cobaltdeficient diets, clinical signs are accompanied by a fall of as much as 90% in the vitamin B12 content of the feces. Oral dosing with cobalt resolves the clinical signs, and vitamin B12 levels in the feces return to normal. Parenteral administration of cobalt has no appreciable clinical effect, although some cobalt does enter the alimentary tract in the bile and leads to the formation of a small amount of cobalamin. The essential defect in cobalt-deficient ruminants is an inability to metabolize propionic acid. A key biochemical pathway for

Nutritional Deficiencies

VetBooks.ir

propionic acid from rumen fermentation involves adenosyl cobalamin, one of several cobalt-containing coenzymes of the vitamin B12 complex that is required for the conversion of methylmalonyl coenzyme A to succinyl coenzyme A, both intermediates in the utilization pathway of propionate.1 The propionate-succinate pathway is the first ratelimiting pathway in vitamin B12 deficiency,2 and the lack of vitamin B12 causes accumulation of methylmalonic acid, which can be measured in the serum. The clinical and pathologic signs of cobalt deprivation are preceded by characteristic biochemical changes in tissues and fluids of the body. As soon as depletion begins, the concentration of both cobalt and vitamin B12 decreases in the rumen fluid. Serum vitamin B12, an estimate of the amount of this vitamin in transit, also shows an early decline reflecting reduced rumen synthesis. Serum vitamin B12 declines before liver vitamin B12, confirming that the liver does not serve as an active storage pool. Plasma methylmalonic acid is elevated (>5 µ mol/L) within 35 days of a cobaltdeficient diet being fed, before any reduction in feed intake or live weight occurs, and rumen succinate concentration is elevated after feeding a deficient diet for 6 days.3 In beef cattle, a prolonged moderate cobalt deficiency (83 µg/kg) for 43 weeks results in impaired growth and changes in lipid metabolism, including accumulation of plasma homocysteine and a marked increase of iron and nickel in the liver. The mechanism through which cobalt prevents staggers in sheep grazing pasture dominated by phalaris (Phalaris tuberosa), and possibly canary grass (Phalaris minor) or rhompa grass, a hybrid Phalaris spp., is unexplained. The pathogenesis of ovine white liver disease is unclear, as it is not known if the disease is a simple cobalt deficiency or a hepatotoxic disease in cobalt/vitamin B12– deficient lambs. A cobalt-deficient diet is essential for the development of the disease, which is characterized by hepatic dysfunction and elevated liver enzymes (alkaline phosphatase and aspartate aminotransferase).4 Affected lambs have elevated serum levels of copper compared with cobalt/ vitamin B12–supplemented lambs grazing the same pastures, and dosing affected lambs with copper-oxide needles can induce toxic levels of liver copper. It is suggested that the disease is a manifestation of B12 deficiency exacerbated by factors triggering early hepatic fatty change, resulting in more severe liver damage and loss of intracellular homeostasis, rendering the hepatocytes more vulnerable to other elements such as copper. It is proposed that elevated concentrations of fructans in pasture could also contribute to the pathogenesis of the liver lesion by initiating hepatic lipodystrophy and hepatic insufficiency, and hence reduced growth and ovine white liver disease. The condition

responds to treatment with parenteral vitamin B12. The pathologic changes in lambs grazing cobalt-deficient pastures are related to blood concentrations of vitamin B12, methylmalonic acid, and homocysteine, and lesions are confined mainly to the liver and brain. Acute and chronic hepatitis are characteristic, and the liver lesions are associated with polymicrocavitation of the brain. Hepatic encephalopathy associated with cobalt deficiency and white liver disease has been described in lambs. Symmetric vacuolation and status spongiosus of the neuropil in the brain is seen, with hyperammonemia secondary to the hepatic lesion considered the cause of the brain lesions. Caprine hepatic lipidosis has been induced experimentally using low intakes of low levels of dietary cobalt. Goats provided with a diet that contains the minimum daily requirement of cobalt as specified for sheep not only developed a syndrome characterized by reduced weight gains, dry scruffy hair coat, and a decline in erythrocyte indices, but also lesions consistent with hepatic lipidosis. Goats fed diets containing levels of cobalt less than 0.1 mg/kg DM could experience even greater clinical and pathologic consequences. In moose sickness, there are low concentrations of cobalt and vitamin B12 in the liver and elevated methylmalonic acid in plasma.

CLINICAL FINDINGS

No specific signs are characteristic of cobalt deficiency. A gradual decrease in appetite is the only obvious clinical sign, accompanied by lacrimation, loss of body weight, emaciation, and weakness, often in the presence of abundant green feed. Pica is likely to occur, especially in cattle. There is marked pallor of mucous membranes and normocytic normochromic anemia, and affected animals are easily fatigued. Growth, lactation, and wool production are severely retarded, and the wool may have reduced staple strength (be “tender” or broken). In sheep, severe lacrimation with profuse outpouring of fluid sufficient to mat the wool of the face is one of the most important signs in advanced cases. Signs usually become apparent when animals have been on affected areas for about 6 months, and death occurs in 3 to 12 months after the first appearance of illness, although severe wasting may be precipitated by the stress of parturition or abortion. Cobalt deficiency in pregnant ewes can result in decreased lambing percentage, increased percentage of stillbirths, and increased neonatal mortality. Lambs from deficient ewes are slower to start sucking, have reduced concentrations of serum colostral immunoglobulins, and have lower serum vitamin B12 and higher methylmalonic acid concentrations than lambs from cobalt-adequate dams. Ova recovered from ewes fed a cobalt-deficient diet (0.06 mg/kg DM) are of

819

inferior morphologic grade compared with those from ewes that were supplemented with cobalt, and lambs born from cobaltsupplemented embryo donors had higher serum B12 and were more active within 3 days of birth.5 Moose sickness in Nova Scotia is characterized by a loss of fear of humans, weakness and a staggering gait, apparent blindness, drooping of the ears, emaciation, and infestation by ticks. A decreased intake of food, increasing lethargy, and collapse, accompanied by loss of use of one or more limbs, precedes death.

CLINICAL PATHOLOGY Biochemical Criteria to Determine Cobalt and Vitamin B12 Status Changes in the concurrent serum concentrations of methylmalonic acid and vitamin B12 of ewes and their lambs on cobalt-deficient pastures, and their response to cobalt supplementation, can be evaluated and monitored.1,2 Changes in these measures can be correlated with live-weight gains after the supplementation of lambs from suckling until after weaning. A growth response to cobalt or vitamin B12 supplementation is expected when cobalt levels in herbage fall below 0.08 to 0.1 mg/K DM. Serum and Liver Cobalt and Vitamin B12 Concentrations Serum cobalt concentrations of normal sheep range from 1 to 3 µg/dL (0.17 to 0.51 µmol/L), whereas in deficient animals these are reduced to 0.03 to 0.41 µmol/L. Clinical signs of cobalt deficiency in sheep are associated with serum vitamin B12 levels less than 0.20 mg/mL. This is the standard laboratory test for cobalt status in sheep, with levels of 0.2 to 0.25 µg/L indicative of deficiency, which rapidly increase to 0.5 to 1.0 µg/L following treatment. Depriving sheep of feed for 24 hours results in a marked increase in serum vitamin B12. The serum vitamin B12 levels of sheep at pasture are unreliable indicators of liver vitamin B12. In cattle, serum vitamin B12 values greater than 0.2 µg/L are indicative of normal cobalt nutrition. However, there is considerable variability in this measure between laboratories because of binding within plasma.1 Consequently, liver vitamin B12 is preferred, although the cost of obtaining a liver biopsy restricts the utility of this test. A range of 75 to 250 nmol/kg fresh weight indicates marginal cobalt nutrition in grazing cattle, but this may be too low for cattle on predominantly grain diets.1 Fatty infiltration of the liver will lead to underestimates of the liver concentration of vitamin B12. Concurrent Serum MMA and Vitamin B12 Concentrations.  In ewes and nonsuckling lambs, the serum MMA concentration provides a more precise indication

820

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

of responsiveness to vitamin B12 or cobalt supplementation than serum vitamin B12. Serum MMA concentrations greater than 13 µmol/L indicate responsiveness to supplementation, those from 7 to 13 µmol/L indicate a potential but marginal or inconsistent response, and no response is expected at concentrations less than 7 µmol/L. In a study of cobalt-deficient ewes, serum concentrations of vitamin B12 decreased from 250 pmol/L during early lactation to a nadir of 100 pmol/L at peak lactation, at which time MMA concentration had increased to a range of 7 to 14 µmol/L. In this study, supplemented ewes were significantly heavier, and the vitamin B12 concentration in ewe milk and the livers of their lambs was more than doubled. Supplementation of ewe with cobalt bullets appears to protect the growth performance of the lamb for 90 days and to influence the subsequent serum vitamin B12 response in the lamb to vitamin B12 supplementation. On cobalt-deficient farms in New Zealand, serum vitamin B12 and MMA have been compared as indices of cobalt/vitamin B12 deficiency in lambs supplemented with either cobalt bullets or short- or long-acting preparations of vitamin B12. Serum MMA concentrations greater than 9 to 14 µmol/L were a more reliable indicator of cobalt deficiency, but there is considerable variation between farms. An evaluation of serum MMA and vitamin B12 concentrations used to assess cobalt deficiency in New Zealand found that the reference ranges for vitamin B12 responsiveness may be conservatively high. This could result in an overdiagnosis of vitamin B12 deficiency as a cause of illthriftiness of sheep, and so response trials to assess weight gain following supplementation may be a better alternative. Liver Cobalt.  In lambs, normal liver cobalt ranges from 0.03 to 0.1 µg/g wet weight (WW). Concentrations less than 0.02 µg/g WW (0.07 µg DM) are associated with clinical deficiency, with 0.015 µg/g WW (0.05 µg DM) considered a critical level, and less than 0.025 µg/g WW in a sheep flock is considered marginal. In lambs with clinical signs of ovine white liver disease, mean hepatic cobalt concentrations range from 0.013 to 0.024 µg/g WW. Serum Methylmalonic Acid Because of difficulties with the interpretation of serum vitamin B12 results, other biochemical tests, especially MMA in plasma and urine, are now used. An elevated plasma concentration of MMA is a comparatively early indicator of functional vitamin B12 deficiency, and thus this test can identify a cobalt deficiency earlier. The upper limits of MMA for grain- and pasture-fed animals are 10 and 5 µmol/L, respectively. In cattle, serum MMA less than 2µmol/L is considered normal, 2 to 4 µmol/L marginal,

and greater than 4 µmol/L indicative of a deficiency. Urinary MMA in cobalt-deficient animals is abnormally high, and thus this is also an appropriate test for deficiency. Cobalt-replete lambs have plasma MMA levels less than 5 µmol/L, urinary MMA less than 120 µmol/L, and urinary MMA/creatinine values less than 0.022 µmol MMA/ mmol. An unequivocal result for methylmalonic acid is a concentration of greater than 30 µg/mL for 10 animals selected randomly from a flock. Samples must be acidified to avoid degradation of the methylmalonic acid if they are to be kept for more than 24 hours before testing.

The concentration of vitamin B12 in the liver of cobalt-deficient lambs is 0.1 mg/kg compared with around 0.3 mg/kg in normal lambs. In cattle, concentrations greater than 0.3 mg/kg are necessary for optimum growth, normal levels range from 0.70 to 1.98 mg/kg, and clinical signs occur when liver vitamin B12 is less than 0.1 mg/kg. After oral dosing, the concentration of cobalt in the liver rises but then returns to the pretreatment level in 10 to 30 days. Serum B12 levels reflect cobalt status, and thus it is often useful to submit sera from surviving herdmates when attempting to confirm a diagnosis of cobalt deficiency.

Formiminoglutamic Acid Neither MMA nor formiminoglutamic acid is a normal constituent of urine, and so their presence in urine, without the need for a quantitative measurement, is an indication of cobalt deficiency. In lambs in the later stages of cobalt deficiency, when there is weight loss and ill-thrift, the concentration of urinary formiminoglutamic acid increases to 0.08 to 20 µmol/mL, then rapidly returns to zero following treatment. However, there is little or no increase in the urine of animals with subclinical cobalt deficiency, and thus this measure is not useful as a diagnostic test.

Samples for Confirmation of Diagnosis • Toxicology—50 g liver (ASSAY [Co]), 2 mL serum (ASSAY [B12]) • Histology—formalin-fixed liver (LM)

Hematology Affected animals have normocytic and normochromic anemia, but hemoglobin and erythrocyte values are often within the normal range because of hemoconcentration. There is a decrease in cellularity of the bone marrow that does not respond quickly to the administration of vitamin B12 or cobalt. Affected animals are also hypoglycemic (60%). Clinical disease develops between 9 and 22 months of age. Animals are normal until weaning but then lose weight, develop rough hair coats, and lose incisor teeth. The skeletal changes in hemochromatosis are a result of abnormal bone development. Bone analysis reveals iron levels in affected animals may be 30 to 50 times greater than normal and a decreased percentage of ash in the outer cortex. Periosteal dysplasia and osteopenia are responsible for the pathologic fractures and tooth loss. At necropsy, there is emaciation, firm dark-brown livers and lymph nodes, soft bones, and brown-colored small intestine.

838

Chapter 11 

■ 

Diseases of the Hemolymphatic and Immune Systems

VetBooks.ir

The major histologic changes are hepatocellular siderosis and periportal bridging, along with perivenular fibrosis. Heavy deposits of iron in the liver and deposits of hemosiderin are visible in liver tissue obtained by biopsy. Hepatic iron concentrations in clinically affected cattle range from 1500 to 10500 wet weight (reference range for cattle = 30 min) can develop atelectasis of the down lung that can mimic pneumonia radiographically. Ventrodorsal views assist with localizing lesions in foals and calves. Radiographic evidence of lung disease is common in ill neonatal foals (74% having such lesions in one study), and is not related to clinical evidence of respiratory disease or dyspnea. The characteristics of lung lesions detected in neonatal foals are associated with likelihood of survival. Guidelines for recognition of pulmonary patterns in foals have been proposed (Table 12-2), and these guidelines are likely to be useful aids for interpretation and

Special Examination of the Respiratory System

VetBooks.ir

Opacity

Area(s) of poorly defined or absent/obscured pulmonary vessels

Pulmonary vessels clearly defined in all areas

Diffuse

Normal or airway inflammation/RAO

Other diagnostic tests necessary

Generalized

Discrete

Localized

Size, number, and location

Fig. 12-1  Decision tree for assessing thoracic radiographs of horses. RAO is recurrent airway obstruction or heaves. (Reproduced from Dunkel et al. 2013.14)

Box 12-1  Differential diagnoses for patterns of abnormalities detected on radiographic examination of the thorax of horses No abnormalities detected (but other clinical signs of respiratory disease are present) • Inflammatory airway disease • Heaves • Summer pasture-associated recurrent airway obstruction • Upper respiratory tract disease Diffuse, localized opacities • Caudodorsal lung • Exercise-induced pulmonary hemorrhage • Iatrogenic following bronchoalveolar lavage (transient) • Focal pneumonia • Cranioventral • Infectious bronchopneumonia • Aspiration pneumonia • Pleuropneumonia Diffuse, generalized opacities • Interstitial pneumonia • Pulmonary fibrosis • Pulmonary edema • Acute respiratory distress syndrome One or multiple discrete opacities • Single • Neoplasia • Pulmonary abscess

description of pulmonary patterns in neonates of other species. Indications for thoracic radiography in horses (and likely in other large animal species) include the following:14 • Mild respiratory disease that is unresponsive to treatment • Severe respiratory distress • Thoracic trauma • Signs of respiratory disease and weight loss or recurrent colic • Suspicion of infectious or aspiration pneumonia • Suspected thoracic or mediastinal mass

• Bacterial or fungal granuloma • Foreign body • Multiple • Pulmonary abscesses • Neoplasia • Disseminated fungal (e.g., aspergillus sp.), bacterial (Rhodococcus equi), or parasitic • Equine multinodular pulmonary fibrosis • Eosinophilic interstitial pneumonia • Idiopathic • Other • Bronchiectasis • Tracheal stenosis • Bullae • Increased vascular pattern • Pneumothorax • Pneumomediastinum • Diaphragmatic hernia • Megaesophagus • Foreign body • Pleural fluid (pleuritic, hemothorax, chylothorax)

857

than 50% of affected foals detected by radiographic examination.15 Many pulmonary diseases do not have lesions that are readily detected on radiographic examination. Failure to detect abnormalities on radiographic examination of the thorax does not eliminate pulmonary disease. Furthermore, radiographically detectable signs of lung disease can persist after the animal has clinical and clinicopathologic signs of recovery or improvement. Bronchography utilizing contrast agents is of value in determining the patency of the trachea and bronchi, but general anesthesia is required to overcome the coughing stimulated by the passage of the tracheal catheter. Using a fluoroscope to determine the location of the catheter tip, the contrast agent can be deposited in each dependent lobe in turn. This technique is used infrequently. Radiographic examination of the trachea can reveal the presence of abnormalities in shape, such as occur with tracheal collapse, or the presence of foreign bodies or exudate. Radiographic examination of the head can identify diseases of the paranasal sinuses, ethmoids and pharynx. Radiographic examination is useful in defining diseases of the guttural pouches and in detecting retropharyngeal abscesses or abnormalities, such as the presence of foreign bodies.

MAGNETIC RESONANCE IMAGING The utility of magnetic resonance imaging (MRI) in large-animal medicine is constrained by the size of the imaging bore on MRI units which limits the size of the animal, or anatomic region, that can be imaged. MRI is useful in diagnosis of diseases of the head of horses and other large animals,16 and the anatomy as visualized on MRI of the head of horses and pigs has been reported.17 The lack of units suitable for examination of large animals precludes routine use of this imaging modality.

COMPUTED TOMOGRAPHY

• Esophageal disease • Suspected diaphragmatic hernia Radiography can assist in the recognition and differentiation of atelectasis and consolidation, interstitial and exudative pneumonias, the alveolar pattern of pulmonary disease, neoplasms, pleural effusions, pneumothorax, hydropericardium, and space-occupying lesions of the thorax. Cardiomegaly, abnormalities of the cranial mediastinum, fractures of ribs, and diaphragmatic hernia can also be detected. Thoracic radiography is not as sensitive as is ultrasonography for detecting pulmonary lesions in foals with Rhodococcus equi pneumonia, with fewer

Examination of the lung through computed tomographic (CT) is very sensitive and specific for lung disease in companion animals and is technically feasible in calves,18 foals,19,20 and small ruminants.21 The technique is useful in the diagnosis of mediastinal disease in foals and correlates well with postmortem estimates of the volume of consolidated lung in experimentally induced pneumonia in calves.22 CT is likely to be useful in evaluation of extent, severity and progression of lung disease in calves. The CT anatomy of the head of horses and foals has been described including detailed anatomy of the guttural pouches and paranasal sinuses.23,24 CT imaging of the nasal cavities and paranasal sinuses of horses is useful in the detection of diseases of these

858

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Table 12-2  Guidelines for radiographic pulmonary pattern recognition in foals Alveolar lung pattern (Vessels not visualized. There is displacement of air from the distal air spaces of the lung leading to a relatively homogeneous increase in soft tissue opacity. Formation of air bronchograms is usually associated with the pattern but is not always present) Absent

The pulmonary vessels are easily seen

Minimal alveolar component (< 10%)

No visualization of vessels in < 10% of the lung field. Usually occurs in conjunction with a moderate or severe interstitial lung pattern

Focal (> 10% to 30%)

No visualization of vessels in 11–30% of lung fields. Air bronchograms might or might not be present within   < 30% of lung fields

Localized (> 30% to 50%)

No visualization of vessels in 31–50% of lung fields. Air bronchograms might or might not be present within   < 50% of lung fields

Extensive (≥ 50%)

No visualization of vessels in ≥ 50% of lung fields. Air bronchograms might or might not be present throughout the entire section of lung field

Interstitial lung pattern (Characterization of the non-air-containing elements of the lungs including blood vessels and bronchi) Normal

Clear visualization of vessels. Borders are well defined

Mild increase

The pulmonary vessels appear slightly ill defined (hazy borders with loss of visualization of the fine vascular structures). Mildly lacy appearance to lung field

Moderate increase

The vessels are ill defined, resulting in moderately lacy appearance and increased opacity of the lung field

Marked increase

Significantly increased opacity; vessel borders are barely recognizable

Bronchial pattern (Characterized by alterations in bronchial wall thickness and density, or in bronchial lumen diameter. Note that periobronchial cuffing is a feature of interstitial not bronchial pattern) Normal

Bronchial structures seen in cross section appear as small, thin-walled hollow rings between paired vessels. The bronchial walls are barely distinguishable when viewed side-on and are not clearly visualized at the periphery of the lung field

Moderate increase

A few thickened bronchial walls evident in cross section (“doughnuts”) at the periphery of the lung fields. Longitudinal sections appear as tram lines reaching two-thirds of the way to the lung periphery

Marked increase

Extensive bronchial thickening might be observed, extending far into the periphery of the visible lung field

From: Bedenice D et al. J Vet Intern Med 2003; 17:876.

structures and of the teeth, pharynx, larynx, and guttural pouches.25,26 For example, CT of the head of horses with suspected ethmoidal hematoma provides information that influences the treatment of approximately twothirds of cases, including identification of bilateral disease and the extent of involvement of the paranasal sinuses (and in particular the sphenopalatine sinus). CT is recommended for patients in which the lesion cannot be viewed endoscopically, when sinus involvement or multifocal disease are suspected, or when the lesion has been unresponsive to treatment.27 The technique is technically feasible in ruminants and pigs, including detection of otitis media, pulmonary and pharyngeal abscesses, and congenital pulmonary anomalies in calves.28-30 CT is useful in pigs for quantitation of the extent of atrophic rhinitis and evaluation of pneumonia, in addition to assessment of body composition.31,32

SCINTIGRAPHY (NUCLEAR IMAGING) The basis of pulmonary scintigraphy is detection at the body surface of radiation emitted from the lungs after injection or inhalation of radioactive substances. The technique has been described in both horses and calves. The technique has limited diagnostic usefulness in large animals because of

the need for availability of appropriate isotopes and detection equipment.33 Furthermore, the large size of adult cattle and horses limits the sensitivity of the technique. The technique has been used to determine the distribution of pharmaceuticals administered by aerosolization and the presence of ventilation-perfusion mismatches. Alveolar clearance can be detected using scintigraphic examination. Currently pulmonary scintigraphy is largely a research tool.

ULTRASONOGRAPHY Ultrasonographic examination of the thorax of farm animals and horses is a very useful diagnostic tool. Ultrasonographic examination of the thorax provides diagnostic information that is not obtained by radiographic examination and is more sensitive than radiography in detecting pulmonary abscesses in foals and is more useful than auscultation in detecting consolidation in lungs of preweaned calves.1,15,34 The widespread availability of portable ultrasound units and the ability to image parts of the thorax using ultrasound probes intended for examination of the reproductive tract of mares and cows makes this a potentially valuable diagnostic aid for both field and hospital-based practitioners. Furthermore, the absence of radiation exposure and the “real-time” nature of images obtained by ultrasonography aid

in frequent assessment and monitoring of abnormalities and performance of diagnostic or therapeutic procedures such as thoracocentesis or aspiration of masses. There are limitations to imaging imposed by aerated lung and the bones of the ribcage. Examination of the thorax is limited by the presence of ribs and aerated lungs because the sound waves used to create ultrasound images are reflected from these surfaces. Ultrasonography cannot reveal lesions of the lungs that are not confluent with the visceral pleura. Imaging windows are restricted to the intercostal spaces, but this impediment can be overcome by scanning through adjacent intercostal spaces and angling of the ultrasound beam. Ultrasonographic examination of the thorax should be performed in a consistent manner that ensures thorough examination of the thorax. Preferences for the pattern of examination differ somewhat among examiners, but one common and successful technique is to scan each intercostal space from dorsal to ventral starting at the 17th intercostal space in horses and the 12th intercostal space in cattle. The ultrasound probe is slowly moved from dorsal to ventral while the examiner studies the images. When one scanning of one intercostal space is completed, the probe is moved to the most dorsal aspect of the next intercostal space, and the examination is repeated. Each side of the chest is

Special Examination of the Respiratory System

VetBooks.ir

examined in this manner. This consistent and thorough examination ensures that no important or localized abnormalities are missed. The examination is performed in adult horses and cattle with the animal standing. The rostral thorax is scanned by pulling the ipsilateral forelimb forward. This is more readily achieved in horses than in cattle. Thorough examination of the rostral thorax might require placing the animal in lateral recumbency. Calves and foals can be examined either standing or in lateral recumbency. Ultrasound examination of the thorax is particularly useful for detecting diseases of the pleura, pleural space, or lung surface. This is in addition to the well-documented utility of ultrasonographic examination of the heart and great vessels (see Ch. 11). The normal ultrasonographic anatomy of the thorax of cattle, horses, and calves has been determined. The following is a partial list of disorders or abnormalities detectable by percutaneous ultrasonographic examination of the thorax of farm animals or horses (excluding cardiac abnormalities): • Excess pleural fluid • Characteristics of pleural fluid (flocculent, bubbles, fibrin) • Extent of pleural fluid accumulation • Localized areas of pleural fluid accumulation • Nonaerated lung (atelectatic, consolidated) • Pulmonary abscesses (must be confluent with visceral pleura) • Intrathoracic masses (thymic lymphoma, cranial thoracic mass, gastric squamous-cell carcinoma) • Pleural roughening (“comet-tail” lesions) • Pneumothorax • Pulmonary hematoma • Exercise-induced pulmonary hemorrhage • Hemothorax • Diaphragmatic hernia • Fractured ribs (especially in neonates). Ultrasonographic examination is more sensitive and specific than radiographic examination in detecting the presence of pleural fluid and is particularly useful in the diagnosis and management of pleuritis in horses and cattle and pneumonia in calves. The extent of pulmonary lesions detected at necropsy correlates closely with the results of ultrasonographic examination of calves with pasteurellosis. Ultrasonographic examination is useful in diagnosis of thoracic diseases of cattle. Ultrasonography can identify pulmonary lesions in horses with infectious viral pneumonia. Ultrasonography is useful in identifying the presence of pleural fluid and guiding thoracocentesis to sample and drain this fluid. Ultrasonographic examination of the larynx and associated structures is useful in identifying recurrent laryngeal neuropathy in horses, arytenoid chondritis, and dynamic

laryngeal collapse.35-37 Sensitivity and specificity for ultrasonographic examination of the larynx for detection of recurrent laryngeal neuropathy was 71% to 79% and 86% to 91%, respectively, compared with dynamic endoscopy.38

LABORATORY EVALUATION OF RESPIRATORY SECRETIONS

859

For isolation of viruses associated with disease of the upper respiratory tract, nasal swabs are satisfactory provided a copious amount of nasal discharge is collected, and the swabs are kept moist during transport to the laboratory. Nasal swabs sometimes contain an insufficient amount of secretion, and certain viral pathogens can become inactivated in transit.

SAMPLING RESPIRATORY SECRETIONS

NASAL LAVAGE

When an inflammatory disease process of the respiratory tract is suspected, the collection of samples of secretions and exudate for microbiological and cytologic examination can be considered. The objective is to obtain a sample uncontaminated with environmental flora, which are common in the upper respiratory tract, and to isolate the pathogen(s) or demonstrate inflammatory cells which may be associated with the lesion. This can be done by the following methods: • Swabbing the nasal cavities or the pharynx • Collection of fluid from the paranasal sinus • Collection of fluid from the guttural pouch of Equidae • Transtracheal aspirate • Tracheal lavage • Bronchoalveolar lavage • Thoracocentesis

When larger quantities of nasal discharge are required for research purposes, nasal washings are usually collected, with the simplest technique being irrigation of the nasal cavities and collection into an open dish. From these samples, it is possible to isolate bacteria and viruses and identify immunoglobulins. The development of immunofluorescent and enzyme-linked immunosorbent assay (ELISA) tests for agents of infectious disease has provided reliable systems for the diagnosis of a variety of virus diseases in the early stages of infection. A technique and apparatus are available that obtain much better samples than the conventional cotton-wool swab provides. A vacuum pump aspirates epithelial cells and secretion from the nasal passage and pharynx. Cell smears are then prepared for microscopic examination and the mucus, and cells are used for conventional microbiological isolation.

NASAL SWAB

PARANASAL SINUS FLUID

A swab of the nasal cavities is a reliable method for the evaluation of the secretions associated with disease of the upper respiratory tract such as infectious bovine rhinotracheitis. However, when attempting to assess the health status of the lungs the nasal swab can be unsatisfactory because microbiological examination usually yields a large population of mixed flora, consisting of pathogens and nonpathogens, which is difficult to interpret. Examination of nasal swabs is only useful when seeking to detect specific pathogens (Strep. equi, influenza) and when the diagnostic tests are directed toward detecting these agents.

NASOPHARYNGEAL SWABS

For more reliable results and to lessen the contamination that occurs with nasal cavity samples, swabs of the laryngeal–pharyngeal area can be collected. A swab in a long covered sheath, of the type used for collecting cervical swabs from mares, is easily passed through the nasal cavities to the pharyngeal area. Important differences exist between the microbial isolates from nasopharyngeal swabs and those from lung tissues, which makes nasal swabs unreliable for diagnosis. For example, at the individual animal level, nasopharyngeal swabs and bronchoalveolar lavage show only moderate agreement; at the group or herd level, the isolation rates of various organisms are similar.

Fluid can be collected from the frontal and paranasal sinuses of most of the domestic large animals. Indications for collection of fluid include the presence or suspected presence of disease of the paranasal sinus. Medications can be administered and infected sinuses lavaged using this approach. Absolute contraindications are few but include failure to be able to adequately restrain the animal. Demonstration of fluid in the paranasal sinuses is aided by radiographic examination of the skull. Fluid is collected by percutaneous centesis of the frontal or maxillary sinus and submitted for cytologic and bacteriologic examination (Gram stain, culture). The procedure begins with restraint of the animal, which can include the induction of moderate sedation by administration of alpha-2 agonists and narcotics, or in cattle restraint in a head gate with the head secured with a halter. Next, the area over the centesis site is prepared aseptically and the skin and subcutaneous tissues are anesthetized with local anesthetic. A stab incision (< 1cm) is made in the skin and subcutaneous tissues. A hole is then drilled into the sinus using a Jacob’s chuck with a Steinmann pin (2- to 4-mm diameter). Only a short (5-mm) length of the Steinmann pin should be exposed by the chuck. The hole is drilled by applying steady pressure and making alternating clockwise and counterclockwise

860

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

movements with the chuck. Entry into the sinus cavity is evident as a sudden release of tension and easy passage of the Steinmann pin. The pin is then withdrawn, and sterile polyethylene tubing is inserted into the sinus cavity. Fluid can be aspirated at this time or, if none is forthcoming, 10 to 20 mL of sterile 0.9% saline or similar fluid can be instilled to the sinus cavity. Some of this fluid may run out the nostril if the animal’s muzzle is lower than the sinus. Complications include injury to adjacent structures, including the infraorbital nerve (trigeminal nerve), nasolacrimal duct or parotid salivary duct near its entrance to the oral cavity at the level of the upper cheek teeth. Hemorrhage is usually minor and selflimiting. Subcutaneous emphysema resolves within days. Cellulitis is a risk, especially for animals with septic processes in the paranasal sinuses. Prophylactic administration of antibiotics should be considered in these cases.

TRACHEOBRONCHIAL SECRETIONS

GUTTURAL POUCH FLUID

Comparison of Tracheal Aspirates and Bronchoalveolar Lavage Fluid Examination of tracheal aspirates and bronchoalveolar lavage fluid yields different, but often complementary, information about the lower respiratory tract. The differences between tracheal aspirates and bronchoalveolar lavage fluid arise because cell populations, and types of cell, differ markedly among segments of airways. There is no correlation between cytologic features of tracheal aspirates and bronchoalveolar lavage fluid of horses, and this is probably the case in other species. Tracheal aspirates are representative of cell and bacterial populations of the large conducting airways (trachea and mainstem bronchi), which can originate in both the large and small conducting airways and the alveoli. Secretions of more distal airways can be modified during rostrad movement, such that fluid in a tracheal aspirate is not representative of processes deeper within the lung. Furthermore, disease localized to one region of the lung can alter tracheal fluid. Examination of tracheal aspirates is useful for detecting inflammation of the large airways and for isolation of microorganisms causing disease in these structures.39,40 There is no good evidence that findings on examination of tracheal aspirates correlate with abnormalities in pulmonary function, although they can correlate with exercise performance (racing).41,42 Tracheal aspirates do not accurately reflect lesions in the lungs of horses, but presence of excess mucus, detected on endoscopic examination, is associated with impaired performance, whereas presence of excess neutrophils is not.41 Bronchoalveolar lavage is useful for sampling secretions in the more distal airways. It provides a sample of secretions that have not been contaminated by upper respiratory tract organisms or secretions before collection, and the sample is therefore

Indications for collection of fluid from the guttural pouches of equids include bacteriologic or polymerase chain reaction (PCR) examination to determine whether the horse is infected by S. equi (the etiologic agent of strangles) or to investigate the suspected presence of other inflammatory or neoplastic disease. The preferred method of collection is during endoscopic examination of the guttural pouch. During this examination, fluid can be collected through a polyethylene tube inserted through the biopsy port of the endoscope. Fluid collected in this manner is potentially contaminated by organisms in the upper respiratory tract, and results of bacteriologic examination should be interpreted with caution. Usually, bacteriologic examination is for the presence of S. equi and demonstration of its presence is all that is required for a diagnosis of infection. Fluid can also be obtained from the guttural pouch by blind passage of a firm catheter, such as a Chambers mare catheter or 10 French dog urinary catheter, into the guttural pouch. This procedure requires some skill, and there is always the uncertainty that one might not have actually manipulated the catheter into the guttural pouch. A third technique involves percutaneous puncture of the guttural pouch just posterior to the ramus of the mandible and ventral to the ear. This technique has the potential to yield fluid that is uncontaminated by organisms from the upper respiratory tract, but it carries with it a high risk of injury to the important vascular and neural structures in and around the guttural pouch (internal and external carotid arteries, pharyngeal branch of the vagus nerve, hypoglossal nerve, and others). Percutaneous sampling of guttural pouch fluid should not be undertaken without careful consideration of the risks and benefits of the procedure.

The collection and evaluation of tracheobronchial secretions is a useful method for assessing lower airway disease and is widely used in the determination of the etiology of infectious pneumonia (viral, mycoplasmal, fungal, and parasitic) or the severity of disease (bronchoalveolar lavage fluid cytology in horses with heaves, exercise-induced pulmonary hemorrhage in athletic horses). It is also used as a tool in evaluating the respiratory health of intensively housed animals, such as in piggeries. Cytologic examination of recovered fluid can provide valuable information about the severity, extent, and etiology of disease of the lower airway. There are two methods of sampling tracheobronchial secretions—aspiration of tracheal fluid or lavage of bronchioles and distal airways. Each sampling method yields fluid of differing characteristics and source, and interpretation of the results of examination of these fluids depends on their source and the method of collection.

assumed to be more representative of small airway and, to a lesser extent, pulmonary parenchymal and alveolar secretions or exudates. Bronchoalveolar lavage is useful in the detection of widespread lung disease but not necessarily in the detection of localized disease. Tracheal aspirates, because they in theory represent a composite sample of secretions from all regions of the lung, are likely to be more sensitive in detecting focal disease, such as a pulmonary abscess. Bronchoalveolar lavage fluid composition correlates well with pulmonary function in horses. There is little agreement in cytologic examination of tracheal aspirates and bronchoalveolar lavage fluid of sick and healthy horses, and this difference probably exists in other species. Typically, the proportion of cells that are neutrophils is much higher in tracheal aspirates than in bronchoalveolar lavage fluid of both horses with heaves and normal horses. Mast cells are detected more frequently, and eosinophils less frequently, in bronchoalveolar lavage fluid than in tracheal fluid of normal horses. Tracheal Aspirates Indications for collection of tracheal aspirates include the need for microbiological and cytologic assessment of tracheal fluids. The primary indication is collection of samples for microbiological diagnosis of infectious respiratory disease.39,43-45 Other indications include detection and characterization of inflammation of the conducting airways. Contraindications include severe respiratory distress, although this is not an absolute contraindication, inability to adequately restrain the animal, and severe, spontaneous coughing. Percutaneous tracheal aspirate collection performed in animals with severe coughing can result in development of severe subcutaneous emphysema as a result of the high intratracheal pressures associated with the early phase of coughing. Most animals in which percutaneous tracheal aspirates are collected subsequently have radiographic evidence of pneumomediastinum. Tracheal aspirates can be collected either by percutaneous puncture of the trachea or through an endoscope passed through the upper airways. The advantage of percutaneous collection of tracheal aspirates is that there is minimal risk of contamination of the sample by upper respiratory tract or oropharyngeal secretions. Microbiological examination of the samples is therefore likely to accurately reflect microbes present in tracheal fluid. Collection of tracheal aspirates through an endoscope markedly increases the risk of contamination of the sample with oropharyngeal fluids, and it compromises the diagnostic utility of culture of the sample. This disadvantage is partially alleviated by the use of guarded catheters inserted through the endoscope. The disadvantage of percutaneous collection of tracheal fluid is that it is invasive, and there is a risk of localized

Special Examination of the Respiratory System

VetBooks.ir

cellulitis and emphysema at the site of puncture. Endoscopic collection is relatively noninvasive and readily accomplished. Percutaneous Transtracheal Aspiration Percutaneous transtracheal aspiration is a practical method that has been used extensively in the horse and is adaptable to cattle, sheep and goats. For the horse, a 60-cm no. 240 to 280 polyethylene tube is passed through a 9- to 14-gauge needle inserted into the trachea between two rings. Commercially prepared kits for performing tracheal aspirates in horses are available that include all catheters and needles required. An alternative to polyethylene tubing is to use an 8 to 10 French male dog urinary catheter inserted through an appropriately sized cannula. The site for insertion of the needle or cannula is at the junction of the proximal and middle one-third of the ventral neck. The horse is usually sedated before insertion of the needle or cannula. The skin site is prepared aseptically, and a short stab incision is made after the area has been anesthetized. The cannula is removed to avoid cutting the tube, and the tube is pushed in as far as the thoracic inlet. Fluid typically pools in the trachea at the thoracic inlet in horses with lung disease (the tracheal lake or pool), and it is this fluid that is aspirated. Thirty to 50 mL of sterile saline (not bacteriostatic saline) is rapidly infused. The catheter or tubing should be rotated until tension is felt on aspiration by a syringe. Fluid is aspirated and submitted for cytologic, microbiological, or other examination. Complications such as subcutaneous emphysema, pneumomediastinum, and cellulitis can occur, which necessitates care and asepsis during the procedure. Sudden movement of the cannula during insertion of the tubing may cause part of the tube to be cut off and to fall into the bronchi, but without exception this is immediately coughed up through the nose or mouth. Endoscopic Sampling of Tracheal Secretions The flexible fiberoptic endoscope can be used to obtain tracheal lavage samples and at the same time visualize the state of the airways. The process is as for rhinolaryngoscopic examination with the addition of passage of a catheter through the biopsy port of the endoscope. Tracheal fluid is then visualized and aspirated through the catheter. The clinical advantages of the endoscopic collection include noninvasiveness, visual inspection of the airways, guidance of the catheter, and speed. The use of an endoscope with a guarded tracheal swab minimizes contamination by oropharyngeal secretions but does not eliminate it. Assessment of Results Microbiological examination can yield any one or more of a variety of bacteria,

depending on the species examined, the animal’s age, and its clinical condition. Tracheal aspirates of normal animals rarely yield any bacterial growth on culture. Growth of unusual organisms or known oropharyngeal commensal bacteria from samples obtained by endoscopic examination should not be given undue clinical significance as they probably result from contamination of the tracheal aspirate during collection. Pseudomonas spp. and anaerobes isolated from tracheal aspirates collected by endoscopy are almost always contaminants and of no clinical significance. The extent of contamination of tracheal aspirate samples by oropharyngeal bacteria can be estimated from the number of squamous epithelial cells in the sample. There is an apparent approximate linear relationship between the number of squamous cells per milliliter of fluid and the number of colony-forming bacterial units in tracheal aspirates. Samples containing over approximately 10 squamous epithelial cells per milliliter of tracheal aspirate had markedly greater bacterial contamination. Examination of Gram-stained smears of tracheal fluid is specific but not very sensitive for detection of bacteria, compared with culture. In other words, if examination of a Gramstained smear of tracheal fluid reveals bacteria, then the sample is likely to yield bacteria on culture, whereas failure to detect bacteria predicts poorly the likelihood of growth of bacteria on culture of the sample. This indicates that examination of Gram-stained samples of tracheal fluid does not reliably predict bacterial isolation, and if an infectious etiology is suspected, the fluid should be cultured. Results of the microbiological examination of the tracheal fluid should be consistent with the animal’s clinical condition and expected isolates. Cytologic examination of tracheal fluid is an important diagnostic tool. Various stains are available to aid identification of cell types and numbers in tracheal aspirates. Neutrophils, macrophages, lymphocytes, and epithelial cells are readily identified on the basis of their classical morphology and staining using fast Romanowsky stain (Diff-Quik), but this stain is not suitable for identifying mast cells in equine tracheal fluid and probably that of other species. Leishman’s stain is useful to identify mast cells. Clinically normal horses typically have fewer than 20% to 30% of cells as neutrophils with the majority of remaining cells being macrophages, lymphocytes and epithelial cells. Animals with inflammation of the airways typically have increased cell counts and proportion of neutrophils and large amounts of mucus. Horses with inflammatory airway disease such as heaves typically have more than 20% of the cells as neutrophils (see following “Heaves” section), and those with infectious pneumonia often have 50% to 90% of cells as neutrophils. Exercise markedly increases the proportion of neutrophils in tracheal

861

fluid collected within 1 hour of the horse completing intense exercise.46 The presence of eosinophils is considered abnormal and is consistent with parasite migration (Parascaris equorum in foals, Dictyocaulus viviparus in calves). The presence of hemosiderinladen macrophages is evidence of prior pulmonary hemorrhage. Bronchoalveolar Lavage Bronchoalveolar lavage provides a sample of secretions and cells of the distal airways and alveoli, referred to as bronchoalveolar lavage fluid. It is a widely used procedure in horses and, to a lesser extent, cattle and calves, sheep, camelids,44 and pigs. The procedure can be performed on foals, either sedated or anesthetized with improved fluid recovery in the latter.43 Analyses performed on bronchoalveolar lavage fluid include measurement of cell number and concentrations of various acute-phase proteins, analysis of type of immune proteins and surfactant, culture (usually in pigs and cattle), and use of PCR to detect specific pathogens (e.g., the causative agent of ovine pulmonary adenocarcinoma).47,48 It is a relatively noninvasive procedure that allows cytologic and biochemical evaluations of the lower airways and alveoli, which are useful diagnostic aids when evaluating animals with lung disease. Although fiberoptic bronchoscopy and tracheal aspirates permit assessment of the major bronchi and upper airways, bronchoalveolar lavage offers an extension of the diagnostic potential by sampling the terminal airways and alveolar spaces. The primary indication for collection of bronchoalveolar lavage fluid is acute or chronic lung disease. This includes both infectious and noninfectious diseases, although interpretation of samples collected by passage of the collection tube through the nostrils or mouth is complicated by the inevitable contamination of the sample by oropharyngeal commensal bacteria. Despite this shortcoming, the technique has been used to detect pneumonia associated with Mycoplasma sp. in cattle. Contraindications are few, with respiratory distress being an obvious one. Complications of bronchoalveolar lavage are also few, and include a mild neutrophilia in lavaged sections of lungs and changes in phagocytic function of pulmonary macrophages, and microbial content, for several days after the procedure. Transient bronchial collapse can occur during the procedure in horses and is an indication of airway inflammation.49 A shortcoming of bronchoalveolar lavage is that it lavages only a small region of the lung, with the risk that focal lung disease is not detected. There is clear evidence that important differences can exist in bronchoalveolar lavage fluid from left and right lungs and that the ideal technique involves col­ lection of fluid from both lungs.50 This is best exemplified in pneumonia in horses, in

862

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

which bronchoalveolar lavage fluid from pneumonic horses can contain large numbers and a high proportion of neutrophils or can be normal, depending on the lung or area of lung lavaged. Therefore the bronchoalveolar lavage procedure is a very specific but not very sensitive test for pneumonia in horses. Abnormal lavage fluid is helpful diagnostically, whereas normal results do not exclude the presence of foci of pulmonary disease. The lavage samples may be normal in horses affected with pneumonia or pleuropneumonia, and because of these false-negative results, this is not the best diagnostic technique to evaluate a horse with pneumonia. In contrast, the tracheobronchial aspirates are more sensitive and most horses with pneumonia have cytologic abnormalities. Endoscopic Bronchoalveolar Lavage Endoscopic bronchoalveolar lavage has the advantage of permitting visual examination of the airways during the procedure and selection of the region of the lung to be lavaged. This technique does require access to sophisticated endoscopic equipment. The technique described here for horses can be modified for use in other species. Horses for bronchoalveolar lavage should be appropriately restrained. Sedation is usually essential and is achieved by administration of alpha-2-agonists. Coadministration of narcotics is recommended by some authorities to reduce the frequency and severity of coughing. Butorphanol tartrate 10 mg for a 400-kg horse is recommended, although this drug is not as effective as intratracheal lidocaine at reducing the frequency or severity of coughing when combined with detomidine for collection of bronchoalveolar lavage fluid. Effective suppression of coughing during collection of bronchoalveolar lavage fluid can be achieved by instillation of lidocaine (60 mL of a 0.7% solution—made by diluting 20 mL of 2% lidocaine solution by addition of 40 mL of isotonic saline). The lidocaine solution is administered as the endoscope enters the rostral trachea. A twitch can be applied to the nares. The endoscope must be at least 2 m in length and the external diameter should be 10 to 15 mm. Endoscopes of 10-mm diameter will pass to about the fifth-generation bronchi, whereas endoscopes of larger diameter will not pass quite as far into the lung. The endoscope is passed until it wedges, and then 300 mL of warmed (to reduced bronchospasm) isotonic saline is introduced in 5 × 60 mL aliquots. Air is infused after the last aliquot to ensure that all fluid is instilled. After the horse has taken between one and three breaths, the fluid is withdrawn and the aliquots are mixed. There is no difference in the cytologic composition of the first and subsequent aliquots. Blind Bronchoalveolar Lavage Commercial bronchoalveolar lavage tubes are available for use in horses, and are

suitable for use in adult cattle and calves. The tubes are made of silicone and are therefore considerably more pliable than stomach tubes (which are not suitable for this procedure). The tubes are 2 m in length and have an external diameter of about 8 mm. The horse is restrained and sedated as for endoscopic bronchoalveolar lavage, and the tube is passed through one nostril into the trachea. The tube is then advanced until it wedges, evident as no further insertion of the tube with mild pressure. Continued vigorous attempts to pass the tube can result in the tube flexing in the pharynx and a loop of the tube entering the mouth. After the tube wedges, the cuff on the tube is inflated to prevent leakage of fluid around it, 300 mL of warm isotonic saline is instilled, the tube is flushed with air, and fluid is aspirated. The fluid should be foamy and, if cell counts are high, slightly cloudy. Bronchoalveolar lavage can be performed in conscious sheep by insertion of 1.7-mm external diameter polyethylene tubing through a cannula inserted percutaneously in the trachea. The tubing is inserted until resistance is detected (about 40-45 cm in an adult sheep) and the lung is lavaged with 30 mL of sterile isotonic saline. Laboratory Assessment of Tracheobronchial Secretions A problem with comparison of cell counts of bronchoalveolar lavage fluid reported by different authors is the use of inconsistent quantities of fluid to perform the lavage. The use of different volumes alters the extent of dilution of the fluid. There is a need for uniformity in technique. An approach to this problem has been to measure substances in the bronchoalveolar lavage fluid that can provide an indication of the extent of dilution of the sample. Both endogenous (urea, albumin) and exogenous (inulin, methylene blue) markers have been used. Dilution factors using urea concentration in plasma and in bronchoalveolar lavage fluid appear to be useful. The assumption is that urea concentrations in bronchial and alveolar secretions will be identical to that in plasma. The formula for correcting for dilution that occurs during collection of bronchoalveolar lavage fluid is: Dilution factor = Urea concentration in bronchoalveolar lavage fluid Urea concentration in plasma where urea concentration in bronchoalveolar lavage fluid and in plasma is expressed in the same units. The volume of the pulmonary epithelial lining fluid can then be calculated: Pulmonary epithelial lining fluid volume = Dilution factor × Volume of bronchoalveolar lavage fluid Samples for cytology are submitted for preparation involving centrifugation of the

sample to concentrate cells for preparation of slides for staining and microscopic examination. At least for samples from horses, examination of smears made directly from the sample, without centrifugation, is diagnostically useful. As for tracheal fluid, the proportion of mast cells in equine bronchoalveolar lavage fluid is underestimated if cells are stained with fast Romanowsky stain (DiffQuik). Ideally, five fields are examined for each slide, rather than simply counting 400 cells, to ensure that the cell proportions are accurately reported, particularly for mast cells.51 Diagnostic Value The aspirates from normal animals contain ciliated columnar epithelial cells, mononuclear cells, and a few neutrophils with some mucus. Bronchoalveolar lavage fluid samples can be collected at 24-hour intervals without affecting the composition of the fluid, whereas collection as soon as 2 hours can result in a neutrophilic response.45 The concentration of the cells depends on the volume of fluid infused and the disease status of the animal. Representative values for various species are listed in Table 12-3. The general pattern is that animals with inflammatory airway disease, either infectious or noninfectious, have a higher proportion of neutrophils than do disease-free animals. However, ranges of normal values vary considerably depending on the species, the age of the animal, and its management (primarily housing conditions). Care should be taken not to overinterpret findings on examination of tracheal aspirates or bronchoalveolar lavage fluid. Although there is good correlation between microbiological results and cell counts in bronchoalveolar lavage fluid of calves with pneumonia and Thoroughbred racehorses with inflammatory airway disease, this association might not hold for all diseases or species. There is the potential for a seasonal effect on bronchoalveolar lavage fluid composition, with mastocytosis occurring more commonly in the antipodean spring and neutrophilia and eosinophilia more common in the summer.52 Aged horses have a higher percentage of lymphocytes and lower proportion of macrophages than do younger horses.53 The clinical importance of this finding is unclear. Thoracocentesis (Pleurocentesis) Paracentesis of the pleural cavity is of value when the presence of pleural fluid is suspected and, in the absence of ultrasonographic examination, needs to be confirmed, and when sampling of pleural fluid for cytologic and bacteriologic examination is indicated. The primary indication for sampling pleural fluid is the presence of excess pleural fluid. Sampling of pleural fluid is usually accompanied by therapeutic drainage, in which case the cannula used for sampling is

Special Examination of the Respiratory System

863

VetBooks.ir

Table 12-3  Representative results of cytology of bronchoalveolar lavage fluid of cattle, sheep, pigs, and horses Total nucleated cell count (cells × 109/L)

Neutrophil (%)

Macrophages (%)

Lymphocytes (%)

Eosinophils (mast cells) (%)

Species

Disease status

Volume infused (mL)

Weaner pigs

Normal

15–30

0.7 ± 0.2

2.0 ± 1.2

95.6 ± 2.7

1.7 ± 1.2

NR

Weaner pigs

Respiratory disease

15–30

0.9 ± 0.3

7.0 ± 4.2

87.9 ± 5.9

3.7 ± 2.0

NR

Adult sheep

Normal, pastured

30

NR

6.9 ± 5.8

81.1 ± 15.3

10.8 ± 15.8

1.2 ± 2.7

Adult sheep

Normal, housed

30

NR

21.8 ± 23.4

57.6 ± 19.6

16.1 ± 12.6

4.5 ± 9.5

Adult sheep

Respiratory disease

30

NR

26.8 ± 16.8

55.4 ± 20.9

11.6 ± 11.1

6.2 ± 8.6

Calves (2–3 months old)

Normal

240

NR

12 ± 10

86 ± 10

2±1

0

Calves (2–3 months old)

Parasitic (Dictyocaulus viviparus) pneumonia

240

NR

20 ± 20

20 ± 10

2±1

70 ± 10

Cattle (6–10 months old)

Healthy

180–240

1.4 ± 0.3

32° C, >90 F) or moderate (21° C, 69 F) ambient temperatures with an intranasal vaccine.9 The preexistence of some local antibody from natural exposure or coinfection with a virulent strain of the virus may also restrict the multiplication of the vaccine virus, especially the temperature-sensitive mutants. Temperature-Sensitive BHV-1 Modified Live Vaccine.  An intranasal BHV-1 vaccine containing an MLV strain whose growth is restricted to the upper respiratory tract has been developed in Europe. The vaccine strain is chemically treated to produce a temperature-sensitive characteristic, so that it cannot replicate at the body temperature of the animal. Prebreeding vaccination of replacement heifers with the vaccine provides fetal protection. The vaccine is efficacious and safe for use in pregnant cattle. Intranasal vaccination stimulates both systemic and local cellmediated immunity and antibody. Disadvantages of Modified Live Vaccines.  The extensive use of MLV vaccines has reduced the incidence of clinical disease but there are some potential disadvantages. MLV vaccines must be stored and handled properly to avoid loss of potency. The parenteral MLV vaccine is potentially abortigenic and cannot be used on nonimmune pregnant cattle. The virus in MLV vaccines can also become latent following vaccination. Fatal, generalized BHV-1 infection has been associated with vaccination of beef calves under 3 days of age with MLV containing BHV-1 and PI-3. An outbreak of meningoencephalitis occurred in purchased Holstein Friesian male calves vaccinated intranasally at 1 and 3 weeks of age with a commercial MLV vaccine containing BHV-1, bovine virus diarrhea virus (BVDV), PI-3, bovine adenovirus infection type-7 and bovine respiratory syncytial virus (BRSV). Parenteral vaccination was recommended as the proper vaccination protocol. The isolated virus was classified as BHV-1.1. Shedding of Virus by Vaccinated Animals.  There is some concern that MLV-vaccinated calves may shed the vaccine virus, which could then spread to pregnant cattle, resulting in abortion. In calves vaccinated with the intranasal vaccines, the virus replicates in the respiratory tract and is shed for 7 to 14 days. In nonimmune calves, replicating virus can

Diseases of the Bovine Respiratory Tract

VetBooks.ir

be detected 9 hours after vaccination, with peak shedding occurring at 4 days. However, the intranasal vaccination of feeder calves at 7 months of age does not result in transmission of the vaccine virus to nonvaccinated animals comingled with vaccinated calves. Calves vaccinated with a live temperature-sensitive mutant of BHV-1 vaccine were protected against clinical illness from experimental challenge, but excreted the virus 2 months later following treatment with corticosteroids. This emphasizes the general principle that the use of a MLV vaccine implies a continuing commitment to vaccination that may reduce the incidence of disease but is unlikely to eradicate the infection. Inactivated Vaccines.  Inactivated virus vaccines were developed because of some of the disadvantages of MLV vaccines. The vaccines contain high levels of inactivated virus or portions of the virus particle (glycoproteins) supplemented with an adjuvant to stimulate an adequate immune response. Inactivated vaccines are given intramuscularly or subcutaneously. They do not cause abortion, immunosuppression, or latency, although they do not prevent the establishment of latency by field strains. They do not cause shedding and are safe for use in and around pregnant animals. They are also relatively stable in storage. Inactivated vaccines, however, may not be as efficacious as MLV vaccines because of the potential for destruction of some of the protective antigens during the inactivation process. They require two doses of the vaccine and protection is not observed until 7 to 10 days following the second dose of the vaccine, which is usually given 10 to 14 days after the primary vaccination. A major disadvantage of both the MLV and inactivated vaccines is that neither allows for differentiation between vaccinated and naturally infected animals. These factors render conventional vaccines ineffective for a concurrent vaccination and eradication strategy and inappropriate for use in breeding bulls for export market or artificial insemination units that demand BHV-1-free animals. These limitations, along with major advances in molecular biology and protein purification techniques, have encouraged the development of genetically engineered attenuated vaccines and nucleic acid–free subunit vaccines. Subunit Vaccines.  A subunit vaccine contains only one or more of the antigens of the pathogen necessary to evoke protective immunity, and lacks the components that might cause unwanted side effects. The major surface glycoproteins of the BHV-1 are the antigens responsible for stimulating protective immunity. To produce a subunit vaccine containing only surface glycoproteins, the proteins are isolated from the virus of

virus-infected cells, or the peptides can be synthesized. The major glycoproteins of BHV-1 originally designated gI, gIII, and gIV are now named gB, gC, and gD, and they induce high levels of antibody in cattle that are fully protected from experimental disease. The level of immunity based on serum antibody titers and protection against experimental challenge is much greater with the individual glycoproteins than are those immunized with commercially available inactivated vaccines. BHV-1 subunit vaccines provide a number of advantages: • They do not contain live virus and therefore cannot be shed to other animals, cause abortion, or establish latent infections. • They prevent infection and disease. • They are not immunosuppressive. • Serologic assays, based on one or more antigens not present in the vaccine, provide a potential to differentiate vaccinates from naturally infected animals. Prevention of infection by the use of a BHV-1 subunit vaccine combined with the use of a diagnostic test to identify infected cattle offers the potential for vaccination of breeding bulls for artificial insemination units and export and for eradication of the virus. The potential disadvantages of subunit vaccines include the following: • Because of the amount of glycoprotein needed, two immunizations may be necessary for protection. • Subunit vaccines will have to be compatible with the commonly available multivalent vaccines. • The efficiency of subunit vaccines is highly dependent on the use of an effective adjuvant. Marker vaccines or DIVA (differentiation of infected from vaccinated animals) attenuated or inactivated vaccines are based on deletion mutants of one or more viral proteins, which allows the distinction between vaccinated and infected animals based on respective antibody responses. This vaccine approach was very successful in eradication programs for pseudorabies. A marker vaccine must be accompanied by a diagnostic test, which enables distinction of infected from vaccinated animals. These tests detect antibodies against a glycoprotein that is lacking in the vaccine. The desirable characteristics of the companion diagnostic test include the following: • Antibodies are detectable in 2 to 3 weeks after infection, both in vaccinated and unvaccinated cattle. • Antibodies must persist for at least 2 years, preferably lifelong. • A low level of virus replication gives rise to detectable antibody formation.

959

• Cattle repeatedly given the matching marker vaccine remain negative in the test. • The test should be suitable to detect antibodies in milk. • The test has high sensitivity and specificity in comparison with conventional antibody tests. Mutants of BHV-1 have been developed by deleting one or more of the nonessential glycoproteins. Marker vaccines offer the advantage of evaluating the effect of vaccination on the circulation of the field virus under naturally occurring conditions. Using a gEdeleted BHV-1 strain, both a killed virus and MLV marker vaccine have been developed. These vaccines induce all the relevant immune responses against BHV-1-specific immune reactions, including antibodies against gE. Both vaccines have the capacity to reduce, and even to stop, the spread of BHV-1. A serologic test that detects gE-specific antibodies in serum and milk is also available. These vaccines have been tested according to the current European requirements for the development of bovine vaccines. The live vaccine is safe in pregnant cattle and is considered safe for all kind of breeding cattle, including bulls. The live-virus marker vaccine is also efficacious in the presence of maternal antibody, and vaccination of very young calves, irrespective of their BHV-1 status, can be recommended. An inactivated BHV-1 gE-negative vaccine resulted in only a slight decrease of about 1.4 liters per cow in milk production after a double vaccination. One concern with this use of modified live BHV-1 gE-negative vaccines is the potential for recombination of vaccine-virus and fieldvirus strains resulting in the emergence of virulent BHV-1 virus that is gE-negative on serologic testing. This potential can be mitigated by development of double mutant vaccine strains, such as a gE and thymidine kinase mutant Bo-HV-1 strain.10 Combination or Multivalent Vaccines.  The vaccines available for the control of diseases associated with BHV-1 infection are mostly multivalent antigen vaccines containing other respiratory pathogens such as PI-3, BRSV, and BVDV. Some also contain the antigens for the control of leptospirosis and campylobacteriosis. Vaccines containing only BHV-1 are not in common use. A Canadian field trial to compare the serologic responses in calves to eight commercial vaccines against BHV-1, PI-3, BRSV, and BVDV found some differences. Antibody responses to BHV-1 were higher in calves vaccinated with MLV vaccines than in those vaccinated with the inactivated vaccines. There were no differences in seroconversion rates and titers to BHV-1 between intranasal and MLV IM vaccines following a single vaccination. However, after double vaccination with MLV BHV-1 vaccines, both seroconversion rates and changes in titers to the virus were higher

960

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

in calves vaccinated IM than in those vaccinated intranasally. Whether or not these differences in antibody titers reflect differences in vaccine efficacy against naturally occurring disease in the field situation is unknown. The vaccination of calves with multivalent vaccines containing MLV or MLV and inactivated BHV-1 is associated with virusspecific interferon gamma production and protection from clinical disease as a result of challenge 5 days after a single vaccination. Immunization and Latency.  Immunization with vaccines, as with natural infection, does not prevent subsequent infection and the possibility of latency. Vaccination Programs in Herds Beef Breeding Herds.  Beef calves should be vaccinated 2 to 3 weeks before weaning as part of a preweaning preconditioning program. Calves vaccinated with the parenteral MLV BHV-1 vaccine before colostral BHV-1 antibody titers reach low levels do not develop an immediate, active serologic response, as indicated by serologic titers, but are sensitized to the virus. Revaccination at a later date, when maternal antibodies have decreased to undetectable levels, results in a marked serologic response. Heifer and bull replacements are vaccinated at least 2 weeks before breeding. When outbreaks of the respiratory disease occur in unvaccinated beef herds, all cattle in the herd may be vaccinated with the intranasal vaccine. Whether or not beef herds should be vaccinated annually following the initial vaccination is uncertain. There are field reports of outbreaks of abortion as a result of the virus in beef cattle that were vaccinated 3 years previously, which suggests that revaccination of breeding females every 2 years may be indicated. Because both natural infection and vaccination results in latent infection, it may be that the persistence of the virus, combined with natural exposure, may result in persistence of antibody. The duration of protective immunity following vaccination is uncertain, but usually lasts 1 year. Antibodies last for at least 5.5 years in heifers following experimental infection and complete isolation during that time. The MLV BHV-1 vaccine given intranasally or parenterally can enhance the prevalence of infectious bovine keratoconjunctivitis in beef calves vaccinated between 4 and 10 months of age, when the risk for the ocular disease is highest. The explanation for the pathogenic mechanism is uncertain. Feedlot Cattle.  Feedlot cattle should be vaccinated at least 10 days before being placed in the lot, especially one in which the disease may be enzootic. If this is not done, a high incidence of the respiratory form of the disease may occur in recent arrivals. If vaccination before arrival is not possible, the next best procedure is to vaccinate the cattle

on arrival and place them in an isolation starting pen for 7 to 10 days, during which time immunity will develop. A 2015 metaanalysis concluded that in natural exposure trials, beef calves vaccinated with commercially available vaccines against BHV-1, BVDV, BRSV, and PI-3 had slightly less than half the risk of developing clinical signs of pneumonia and approximately 1/5th the risk of dying from respiratory disease. Moreover, vaccination with modified live or inactivated IBR vaccine decreased the risk of developing clinical signs of respiratory disease by 39% to 46%, respectively, in beef and dairy calves to experimental challenge compared with unvaccinated controls.11 Collectively, this is strong supportive evidence that vaccination against IBR is effective in beef calves in North America. Dairy Cattle.  The necessity of vaccinating dairy cattle will depend on the prevalence of the disease in the area and in the herd and the movement of cattle in and out of the herd. A closed herd may remain free of BHV-1 infection indefinitely and vaccination may not be indicated. But to avoid unpredictable abortion storms as a result of the virus in dairy herds, heifer replacements should be vaccinated for the disease 2 to 3 weeks before breeding. Vaccination of a large dairy herd with a persistent BHV-1 infection has been successful in controlling the respiratory form of the disease. The intranasal vaccine has been used extensively in newborn calves in problem herds, but its efficacy at such an age is unknown. The parenteral vaccination of beef calves under 3 days of age with an MLV BHV-1 and PI-3 vaccine caused high mortality. If the systemic form of the disease poses a threat to a potential calf crop, the pregnant cows could be vaccinated with the intranasal vaccine in late pregnancy; this will increase the level of colostral antibody available to the newborn calf and will provide newborn calves with protection against the highly fatal systemic form of the disease. A 2015 meta-analysis concluded that in natural exposure trials, dairy calves vaccinated with commercially available vaccines against BHV-1, BVDV, BRSV, and PI-3 had similar risk of developing clinical signs of pneumonia and dying from respiratory disease than unvaccinated controls.11 The markedly different effect of vaccination in dairy calves to that seen in beef cattle may be because respiratory disease occurs most frequently before 6 months of age in dairy calves. However, as indicated previously, vaccination with modified live or inactivated IBR vaccine decreased the risk of developing clinical signs of respiratory disease to experimental challenge by 39% to 46%, in beef and dairy calves, respectively, compared with unvaccinated controls.11 Collectively, there is moderate supportive evidence that vaccination against IBR is effective in dairy calves in North America.

Bulls intended for use in artificial insemination centers present a special problem of disease control because the virus in semen can have severe consequences on reproductive performance. Bulls that are seropositive to the virus must be considered as carriers and potential shedders of the virus, and should not be allowed entry to these centers. Not all bulls that are seronegative can necessarily be considered free of the virus, and regular attempts at the isolation of the virus must be made from preputial washing and semen. Bulls that become infected while at the centers should be kept isolated, culled, and replaced with clean bulls. Bulls from herds that routinely vaccinate against BHV-1 should not be vaccinated with conventional vaccines if destined for an artificial insemination center. Cattle destined for export should not be vaccinated in case importing countries prohibit the introduction of seropositive cattle. This will not guarantee that such animals will not become positive from natural infection. The use of marker vaccines has some potential in breeding bulls intended for artificial insemination units and for export. Eradication Eradication of the BHV-1 virus from a single herd or the cattle population in a country can be considered as an alternative to vaccination, particularly when the initial prevalence of infection based on serology is low.2 Serologically positive animals are removed or culled, and only seronegative animals introduced into the herd. Control is focused on segregation and elimination of seropositive animals and reduction of animal movement to prevent spread. This approach is not feasible in countries with extensive cattle populations and where management practices result in movement of cattle from one region to another. Eradication Using Marker Vaccines.  Some countries are beginning an immunization program with the marker vaccines, which will protect the cattle against disease but still allow differentiation between vaccinated animals and those that have been naturally infected and are potential carriers of the latent virus. These infected animals could be eliminated over a period of time. Successful eradication depends not only on the efficacy of the vaccine but also on the quality of the tests. False-positive test results can lead to unnecessary culling of cattle, an increase of costs, and reduced cooperation of farmers in the eradication program. As an example, a compulsory eradication program for BHV-1 began in The Netherlands in 1998. The program required that farms either vaccinate all cattle twice yearly or be approved for a certified BHV-1-free or specific-pathogenfree (SPF) status. To become a certified BHV-1 free herd, cattle have to be sampled individually and all seropositive animals

Diseases of the Bovine Respiratory Tract

VetBooks.ir

culled as soon as their status is known. The BHV-1-free herd status is monitored by monthly bulk milk samples. The spread of BHV-1 between herds can be prevented using a surveillance system of sampling herds annually, both individual milk samples and blood samples. Herds with BHV-2 infected (seropositive) animals are required to vaccinate with a glycoprotein E (gE)-negative BHV-1 vaccine. The vaccine may be either an inactivated or live vaccine both based on a spontaneous BHV-1 mutant without the complete gE gene. These so-called marker vaccines or DIVA vaccines allow the identification of cattle infected with the wild-type BHV-1 within a vaccinated population using a gEELISA or a commercially available gE-blocking ELISA that both specifically detect gE antibodies. The eradication program is based on the presumption that all BHV-1 wild-type strains express gE and induce antibodies that can be measured with a gE-blocking ELISA. Loss of Certification.  The probability of and risk factors for the introduction of BHV-1 into SPF Dutch dairy farms has been examined. A total of 95 SPF dairy farms were monitored for 2 years, during which time 14 introductions of infectious diseases occurred on 13 of the 95 farms, for a total incidence rate per herd-year at risk of 0.09. Outbreaks were usually associated with allowing cattle to return to their farm, cattle grazed more often at other farms, and protective clothing less often provided to the veterinarian. For a successful eradication program, farms should remain BHV-1 free, which can be achieved by a more-closed farming system. A more-closed farming system is one that rules out the possibility of direct contact with other cattle from other farms. Also, the farmer requests that professional visitors such as veterinarians and artificial insemination (AI) technicians to wear protective farm clothing when handling cattle. Protective farm clothing includes coveralls or overcoats and boots that can be worn over “off-farm” clothing and that the farmer provides to the visitors before handling cattle. A sanitary barrier is a covered area outside the barn in which visitors put on protective farm clothing over their off-farm clothes. A sanitary barrier has a “dirty” side, where visitors leave their off-farm boots, and a “clean” side, where visitors wear protective clothing and can enter the barn. All of these measures would be economical. TREATMENT AND CONTROL Treatment Antimicrobial treatment for animals with a fever to address concurrent bacterial pneumonia (see treatment recommendations in this chapter for Mannheimia hemolytica) (R-1)

Control Vaccination of beef calves > 6 months of age against BHV-1 (preferably with modified live glycoprotein E–negative vaccine) or with modified live or killed vaccine against BHV-1, BVDV, BRSV, and PI-3 (R-1) Vaccination of dairy calves less than 6 months of age against BHV-1 (preferably with modified live glycoprotein E–negative vaccine) (R-2) Vaccination of dairy calves less than 6 months of age with modified live or killed vaccine against BHV-1, BVDV, BRSV, and PI-3 (R-3)

FURTHER READING Biswas S, Bandtopadhyay S, Dimri U, Patra PH. Bovine herpesvirus-1 (BHV-1)—a re-emerging concern in livestock: a revisit to its biology, epidemiology, diagnosis, and prophylaxis. Vet Quart. 2013;33:68-81. Graham DA. Bovine herpes virus-1 (BHV-1) in cattle—a review with emphasis on reproductive impacts and the emergence of infection in Ireland and the United Kingdom. Irish Vet J. 2013;66:15. Muylkens B, Thiry J, Kirten P, Schynts F, Thiry E. Bovine herpesvirus 1 infection and infectious bovine rhinotracheitis. Vet Res. 2007;38:181-209. Nandi S, Kumar M, Manohar M, Chauhan RS. Bovine herpes virus infections in cattle. Anim Health Res Reviews. 2009;10:85-98. OIE Terrestrial Manual, Chapter 2.4.13, Infectious bovine rhinotracheitis/infectious pustular vulvovaginitis, 2010. (Accessed 15.19.15, at: .).

REFERENCES

1. Mahony TJ. Vet J. 2010;184:124. 2. Raaperi K, et al. Vet J. 2014;201:249. 3. Tizioto PC, et al. PLoS ONE. 2015;10.1371. 4. Rivera-Rivas JJ, et al. Vet Immunol Immunopathol. 2009;131:167. 5. Grissett GP, et al. J Vet Intern Med. 2015;29:770. 6. Mahajan V, et al. J Comp Path. 2013;149:391. 7. Geraghty T, et al. Res Vet Sci. 2012;93:143. 8. Moeller RB, et al. J Vet Diag Invest. 2013;25:136. 9. Grissett GP, et al. Am J Vet Res. 2014;75:1076. 10. Kalthoff D, et al. Vaccine. 2010;28:5871. 11. Theurer ME, et al. J Am Vet Med Assoc. 2015;246:126.

LUNGWORM IN CATTLE SYNOPSIS Etiology The nematode Dictyocaulus viviparus (the bovine lungworm). Epidemiology Disease seen mostly in dairy calves; immunity develops relatively quickly, but cattle will succumb if exposed to overwhelming numbers of infective larvae while grazing. Signs Coughing, tachypnea, dyspnea. Clinical pathology Characteristic larvae in feces (but not present during all stages of disease); eosinophilia; enzyme-linked immunosorbent assay (ELISA) tests for serum antibodies.

961

Lesions Large volumes of consolidation in diaphragmatic lobes of lung, emphysema, worms up to 8 cm long in bronchi (only in patent phase of disease). Diagnostic confirmation Clinical pathology as noted; at necropsy, distribution of lesions in lungs and demonstration of worms in bronchi. Treatment Eprinomectin, avermectins/ milbemycins and benzimidazoles are active against all parasitic stages of D. viviparus; eprinomectin and avermectins also have a persistent protective effect; levamisole also used. Control Vaccination; early-season anthelmintic prophylactic programs using suitable intraruminal boluses or multiple doses of avermectins/milbemycins; keep susceptible animals off potentially dangerous pasture.

ETIOLOGY

The nematode Dictyocaulus viviparus is the only lungworm of cattle. The disease it causes has many local names, including the following: • Parasitic bronchitis • Verminous pneumonia • Verminous bronchitis • Husk • Hoose Bovine lungworm has a very wide distribution through temperate and cold areas and, depending on climatic conditions and season, can cause serious losses.1 The disease reaches its greatest importance in mild, damp regions of the British Isles and parts of western Europe. D. viviparous is also carried by the European bison, in which it causes a disease with similar pathologic lesions to those in cattle.2,3 Deer carry similar parasites, including D. eckerti and D. capreolus. It is uncertain whether deer play a role in the transmission of D. viviparus but lungworm species are generally host specific.

LIFE CYCLE

Adult lungworms live in the trachea and bronchi. The females are prolific egg producers, and it has been estimated that a single infested calf may contaminate a pasture with 33 million larvae. The eggs are coughed up and swallowed. They hatch in the air passages or alimentary canal, and larvae are passed in the feces. These develop in the dung pat through to the infective third stage, which is protected by cuticles retained from both first and second molts. Because the ensheathed larvae cannot feed, glycogen granules are stored in the intestinal cells. Moisture is essential for the survival and development of the larvae, and a moderate temperature of 18° to 21° C (65–70° F) permits their full development to the infective state in 3 to 7 days. Larvae survive best in cool, damp surroundings, especially when the environment is stabilized by the presence

962

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

of long herbage or free water. Under optimum conditions, larvae may persist for over 1 year. They can overwinter in climates as cold as Canada and Germany. When warmer spring weather arrives, the larvae resume their motility but quickly die once their food stores are depleted. Transmission occurs when cattle ingest third-stage larvae while grazing. These migrate through the intestinal wall to reach the mesenteric lymph nodes. From here they pass via the lymphatics to the venous bloodstream and through the heart to the lungs, where they break into the alveoli. They migrate up the bronchioles to their predilection site in the larger air passages and start to lay eggs some 3 to 4 weeks after infestation. Most adult worms succumb to immune expulsion within a few weeks. These events determine the progression of the clinical syndrome and their approximate timing is as follows: 1. Penetration phase (ingestion to arrival of larvae in lung), days 1 to 7 2. Prepatent phase (larvae in lung), days 7 to 25 3. Patent phase (mature worms in lung) days 25 to 55 4. Postpatent phase (lungworms disappearing from lung), days 55 to 70

EPIDEMIOLOGY

Bovine parasitic bronchitis is a sporadic and largely unpredictable disease. This is because immunity develops more quickly than is the case with many other nematode infections, but nevertheless can remain incomplete for many weeks and may wane in the absence of reinfection. In most grazing seasons, immunity will develop fast enough to protect calves against the accumulating numbers of infective larvae on the grass. The farmer may not even realize that the land is contaminated. Clinical outbreaks occur when weather patterns, management, or other factors result in sudden exposure to a pasture challenge sufficient to overwhelm any immunity that has already developed. In comparison with the gastrointestinal nematodes of cattle, relatively few worms (i.e., a few hundred or thousand) are required to produce clinical signs. Thus the disease is almost entirely confined to grazing cattle and occurs most frequently in young animals in their first year on grass, although outbreaks are becoming more common in adults.4 The epidemiology of lungworm disease is largely concerned with factors determining the number of infective larvae on the pasture and the rate at which they accumulate. Infective D. viviparus larvae are relatively inactive and are incapable of traveling more than 5 cm from the dung pat. Factors that disperse the larvae more widely over the pasture include mechanical spread by the following: • Rain • Earthworms

• Wheeled vehicles • Human and animal feet A fungus, Pilobolus, plays a particularly important role in this process and can transfer larvae across field boundaries. Fungal spores on grass pass through the grazing animal and germinate in the feces. Dictyocaulus larvae climb onto the sporangium (fruiting body), which fills with water and bursts, propelling the fungal spore and the lungworm larvae for distances of up to 3 m.5 Dairy calves are most vulnerable to lungworm disease because they are often reared indoors until 4 to 5 months of age and then placed on paddocks grazed each year by successive calf crops. If the paddocks are heavily contaminated, acute disease may occur in 1 week or so. Usually, however, only sufficient larvae survive the winter to induce lowgrade asymptomatic infections in the susceptible calves, which then start to recontaminate the pasture and recycle the infection. With the high stocking densities commonly used, pasture challenge can reach pathogenic levels within 2 to 4 months. This model does not satisfactorily explain all outbreaks, and it has been suggested that larvae may be washed into the soil to emerge later (e.g., onto hay aftermath). Beef calves at grass with their dams are less likely to be affected as this system provides fewer opportunities for large numbers of larvae to accumulate, but outbreaks can occur particularly after weaning in the autumn.6 In older animals, larvae ingested in the autumn become hypobiotic and resume their development in the following spring. This event occasionally causes disease in housed cattle6 but such infections are usually asymptomatic and provide a source of pasture contamination when these carrier animals are put out to graze. This is thought to be the main source of infection in more severe climates where overwintering larvae may not survive on the pasture, but carrier animals have also been incriminated in disease outbreaks in, for example, Louisiana in the United States. Immunity to reinfestation occurring after initial exposure to D. viviparus is variable in degree and duration. It normally provides protection during the first grazing season and is boosted by exposure to overwintered larvae at the beginning of each subsequent grazing season. Cattle removed from infected pastures for long periods can suffer clinical disease when reexposed. Recently the number of outbreaks of parasitic bronchitis in yearling and adult cattle in the United Kingdom, Denmark and some other countries has been rising. Reasons for this are speculative but include the following: • A decline in the use of vaccination • Changes in weather patterns and management systems • Use of highly effective anthelmintic strategies in the first grazing season

that may prevent adequate antigenic exposure

PATHOGENESIS

Migrating D. viviparus larvae provoke little damage until they reach the lungs. Thereafter, passage of larvae up the bronchioles causes them to become blocked by mucus, eosinophils, and other inflammatory cells, leading to collapse of the alveolae that they supply. Coughing and dyspnea occur if a sufficiently large volume of lung tissue is affected. This is accompanied by pulmonary edema and interstitial emphysema. As no structural damage has yet occurred, treatment at this stage in the disease produces an immediate clinical response. Later, however, when mature parasites are in the major bronchi, eggs and fragments of worms killed by immunity are aspirated and provoke a foreign-body pneumonia. Secondary bacterial infections establish and sequelae such as bronchiectasis occur. Such lesions are slow to resolve, and treated animals will require a long recovery period. Later still, once all or most of the worms have been expelled, the alveolar lining cells of some 25% of recovering animals become cuboidal and nonfunctional. The reason for this is unknown but may be a response to substances released by the dead worms. Because this reaction is irreversible, many animals affected in this way will die. The response of the lung varies widely depending on the number of larvae ingested, the nutritional status and age of the host, and whether or not it is exposed to lungworm infection for the first time. Vaccinated animals or those that have recovered from clinical or subclinical infection may cough and even become tachypneic if grazed on contaminated pasture. This is known as the “reinfection syndrome” and occurs as many larvae reach the lungs before succumbing to the immune response. Exposure of older previously infected animals to massive challenge may invoke a severe or fatal hypersensitivity reaction.

CLINICAL FINDINGS

Outbreaks vary in severity from sporadic coughing with no apparent production loss to acute cases with a rapidly fatal outcome. Individuals within a group are usually affected to varying degrees. Poorly nourished animals appear less able to withstand lungworm infection. Nevertheless, it is not unusual for severe infestations to be fatal in well-fed calves. Acute cases have rapid shallow abdominal breathing of sudden onset that may reach a rate of 60 to 100 breaths/min. There is a frequent bronchial cough, a slight nasal discharge, a temperature of 40 to 41° C (104– 105° F) and a heart rate of 100 to 120 bpm. The animal is bright and active and will attempt to eat, although respiratory distress often prevents this. Progress of the disease is

Diseases of the Bovine Respiratory Tract

VetBooks.ir

rapid, and within 24 hours dyspnea may become very severe, accompanied by mouth breathing with the head and neck outstretched, a violent respiratory heave and grunt, cyanosis, and recumbency. On auscultation, lung consolidation is evidenced by loud breath sounds, and crackles are heard over the bronchial tree. The crackling of interstitial emphysema commences over the dorsal two-thirds of the lung but is never as evident as in less acute cases. Fever persists until just before death, which usually occurs in 3 to 14 days and is greatly hastened by exercise or excitement. The case-fatality rate in this form of the disease is high, probably of the order of 75% to 80%. Subacute disease is more common in calves than the very acute form. The onset is usually sudden, the temperature is normal or slightly elevated and there is an increase in the rate (60-70 breaths/min) and depth of respiration. An expiratory grunt is heard in severe cases and expiration may be relatively prolonged. There are frequent paroxysms of coughing. The course of the disease is longer, 3 to 4 weeks, and auscultation findings vary widely with the duration of the illness and the area of lung involved. In general, there is consolidation and bronchitis ventrally and marked emphysema dorsally. Affected animals lose weight very quickly and are very susceptible to secondary bacterial bronchopneumonia. The mortality rate is much less than in the acute form, but many surviving calves have severely damaged lungs. Consequently, they may remain stunted for long periods, and breathing may be labored for several weeks. Some surviving calves may show a sudden exacerbation of dyspnea around 7 to 8 weeks after the initial onset of disease. In these relapsed cases the prognosis is grave. Adult dairy cattle are usually immune but sporadic outbreaks do occur as a result of waning immunity. Mortality is low but morbidity can be high, with reduced milk yields causing significant economic loss.7-9 Coughing is a constant feature, but other clinical signs are variable and may include dyspnea, nasal discharge, and weight loss.7 Sudden exposure of immune adults to massive challenge can cause severe interstitial pneumonia.

CLINICAL PATHOLOGY

The presence of D. viviparus larvae in feces confirms lungworm infestation, but their absence does not necessarily exclude the possibility of parasitic bronchitis. No larvae will be passed in the early stages of disease when the causal worms are still immature, nor will they be a constant finding when partially immune animals (e.g., dairy cows) succumb to challenge. In general, larvae can be found about 12 days after signs appear (i.e., around 24 days after infestation occurs). They are few in number at first but may become more numerous later.

Enzyme-linked immunosorbent assay (ELISA) tests using adult or larval worm antigen for the detection of D. viviparousspecific antibodies in serum and in milk (including bulk tank milk) have been developed.10,11,12 Care is required with interpretation because antibodies to adult antigen may not be detectable until several weeks after primary challenge and do not correlate with the immune status of the animal. Eosinophilia is a fairly consistent finding but not pathognomonic. An alternative method, if disease is suspected but the lungworms are still in the prepatent stage, is to examine pasture clippings for larvae. This is a laborious procedure because large amounts of herbage (0.5–1 kg) must be used, and the yield of larvae is low.

NECROPSY FINDINGS

Adult D. viviparus are up to 8 cm long and easily seen when the trachea and bronchi are cut open. Worms may also be recovered by lung perfusion. Up to several thousand may be present in severely affected animals. In prepatent disease, however, careful microscopic examination of bronchial mucus is necessary to find larvae. Adult worms may be few or absent if the case is of sufficient duration for immune expulsion to have taken place. In acute cases, morphologic changes include the following: • Enlargement of the lungs as a result of edema and emphysema • Widespread areas of collapsed tissue of a dark pink color • Hemorrhagic bronchitis with much fluid filling all the air passages • Enlargement of the regional lymph nodes Histologically, the characteristic signs are as follows: • Edema • Eosinophilic infiltration • Dilation of lymphatics • Filling of the alveoli and bronchi with inflammatory debris • Larvae in the bronchioles and alveoli In subacute cases, interstitial emphysema is usually gross. Areas of dark pink consolidation are present in the diaphragmatic lobe and may also occur in other lobes. They can occupy two-thirds of the lung volume. There is froth in the bronchi and trachea. The regional lymph nodes are enlarged. Histologically, eggs and larvae can be seen in the air passages, the bronchial epithelium is much thickened, the bronchioles are obstructed with exudate, and the alveoli show epithelialization and foreignbody giant-cell reaction. The reinfection syndrome is characterized by the presence of numerous 5-mm gray-green nodules formed by lymphoreticular cells clustering around dead larvae.

963

DIAGNOSTIC CONFIRMATION

D. viviparus larvae may be demonstrated by placing feces on a fine sieve or dental gauze on the top of a water-filled funnel (the Baermann technique). The larvae that swim into the water and collect at the bottom of the funnel are less than 0.5 mm long, sluggish, and often appear curved or coiled. Their most important diagnostic feature is the presence of easily visible refractile granules in the intestinal cells. Because not all animals will be shedding larvae, samples should be taken from all, or at least a representative proportion, of the group. Grass samples are washed in water with a surfactant and the sediment Baermannized as described previously. A technique that effectively separates larvae from plant debris by migration through agar gel has been reported. Gathering grass close to dung pats maximizes chances of finding larvae. Cattle with parasitic bronchitis are likely to have eosinophilia, and serologic tests can be used to rule out some other respiratory diseases, such as infectious bovine rhinotracheitis (IBR). In view of the uncertainties associated with laboratory tests for parasitic bronchitis and the need for prompt treatment, diagnosis often has to be based on clinical history, signs, and auscultation. Affected animals have usually grazed alongside potential carriers or had access to pasture previously used by susceptible calves or older carrier animals. The timing of the outbreak may coincide with that expected from recycling of an infection initiated by overwintered larva (often 2-4 months after turnout) or recent exposure to heavily contaminated land. Many of the clinical signs of parasitic bronchitis are common to pneumonias of bacterial and viral origin. One feature that may be of value in differentiation is the relative softness and paroxysmal nature of the cough in parasitic infection. DIFFERENTIAL DIAGNOSIS • • • • •

Bacterial bronchopneumonia Acute and chronic interstitial pneumonia Viral pneumonia Acute interstitial pneumonia (fog fever) Heavy infestations with ascarid larvae on pastures contaminated with pig feces

In adult cattle, the major problem in diagnosis is to differentiate the acute form of the disease from acute interstitial pneumonia attributable to other causes. Clinically, the diseases are indistinguishable, and a history of movement onto a new pasture 1 to 2 weeks before the onset of the disease may be common to both. It is necessary to demonstrate D. viviparus antibodies in blood, worms at necropsy, and larvae in the herbage or in the feces of animals that previously grazed the pasture.

964

Chapter 12 

■ 

Diseases of the Respiratory System

TREATMENT

VetBooks.ir

TREATMENT AND CONTROL Treatment Eprinomectin (0.5 mg/kg TOPp, 0.2 mg/kg SC) (R-1) Ivermectin (0.2 mg/kg, SC, PO; 0.5 mg/kg, TOPp) (R-1) Doramectin (0.2 mg/kg, SC, IM) (R-1) Moxidectin (0.2 mg/kg, PO, SC; 0.5 mg/kg, TOPp) (R-1) Albendazole (10 mg/kg, PO) (R-2) Oxfendazole (7.5 mg/kg, PO) (R-2) Febantel (7.5 mg/kg, PO) (R-2) Fenbendazole (5 mg/kg, PO) (R-2) Netobimin (7.5 mg/kg) (R-2) Levamisole (7.5 mg/kg) (R-3) Control Eprinomectin extended-release formulation (1.0 mg/kg, SC) (R-2) Ivermectin long-acting formulation (0.63 mg/ kg, SC) (R-2) Vaccination Live irradiated infective D. viviparus larvae (1000 larvae/calf, PO) (R-2) PO, orally; SC, subcutaneously; TOPp, topical pour on formulation.

Anthelmintics may be used prophylactically to prevent disease from occurring, as a curative treatment once disease strikes, or to prevent reinfection following an outbreak. Avermectins and milbemycins are particularly useful for prophylaxis and prevention of reinfection because they are not only highly effective against the lungworms present in the animals at the time of treatment but have prolonged activity against subsequent incoming larvae. The duration of this persistent effect varies with compound and formulation. Most modern broad-spectrum drugs are active against D. viviparus. Dosage rates and label claims vary from country to country according to local conditions and regulatory requirements. Avermectins and milbemycins (macrocyclic lactones) are particularly potent against immature and mature stages; doses of ivermectin, for example, as low as 0.05 mg/kg, are effective. At commercial dose rates, ivermectin by injection or as a pour-on formulation provides residual protection for up to 28 days; corresponding figures are up to 35 days for doramectin by injection and 42 days both for doramectin as a pour-on formulation and moxidectin by either route of administration. These compounds are given at 0.2 mg/kg by injection and 0.5 mg/kg as a pour-on formulation. Eprinomectin is the compound of choice for adult dairy cattle because it has a nil milk withdrawal period14 and provides residual protection of up to 28 days when given topically (0.5 mg/kg). Albendazole (10 mg/kg),

febantel (7.5 mg/kg), fenbendazole (5 mg/ kg), netobimin (7.5 mg/kg), and oxfendazole (4.5 mg/kg), which are given orally, are active against all stages of the parasite but have no residual activity. Levamisole (oral or injection—7.5 mg/kg; pour on—10 mg/kg) also has activity against lungworm but no persistent effect. Sustained-release intraruminal devices (“boluses”) provide extended periods of protection. For example, fenbendazole is released for up to 140 days from one bolus. There are also pulse release boluses containing oxfendazole that release five or six anthelmintic doses at 3-week intervals. Most boluses normally protect against disease but may allow some worms to establish (in the case of the fenbendazole bolus) or to reach the lungs between pulses (oxfendazole bolus), which may allow immunity to develop. Formulations of eprinomectin extended-release injection have been developed that have greater than 98% efficacy against D. viviparus and provide protection from reinfestation for up 150 days in catlle.13,14,15,16 Similarly, an ivermectin longacting injectable formulation has been shown to have up to 100% efficacy against D. viviparous in cattle for at least 77 days.17 For veterinarians in the field, the outcome of therapeutic treatment is often unpredictable because it depends on the amount of structural damage in the lungs. Best results are obtained early in the course of disease when most pathologic changes can be quickly resolved. In severe cases, treatment may initially exacerbate clinical signs because the death and disintegration of many worms in the air passages releases antigens and adds to the mass of foreign material that can be aspirated. Because of animal welfare considerations and the high risk of mortality, anthelmintic treatments are often combined with an antihistamine or nonsteroidal antiinflammatory drug (NSAID) such as flunixin to reduce the severity of the reaction to the larvae and an antibiotic or sulfonamide to prevent secondary bacterial infection. Severely affected animals should be brought indoors for nursing and all other members of the group removed from the contaminated pasture and placed on clean grazing ground.

CONTROL

Two major strategies of control derive from the premise that the main factor governing the occurrence of disease is the density of D. viviparus larvae on pasture grazed by susceptible cattle. First, cattle grazing potentially contaminated pasture can be protected by vaccination or anthelmintic cover. Alternatively, steps can be taken to ensure that pastures are safe for grazing. This is usually achieved by prophylactic anthelmintic programs, but delaying spring turnout until overwintered larvae have died away is a theoretical option on organic farms.

Sensible grazing management is important in all systems but cannot be relied upon, per se, for controlling parasitic bronchitis in view of the unpredictable nature of the disease. Although natural immunity provides adequate protection on many farms, it cannot be accurately measured nor predetermined. With the possible exception of beef suckler systems, calves should not be run with or follow older cattle because these may harbor asymptomatic patent infections and contaminate the pasture. An important consideration is that clean pasture can be contaminated by larvae from neighboring fields carried on windborne fungal spores (see epidemiology paragraph earlier). Although the numbers of larvae spread in this way are likely to be small, they can initiate the epidemiologic cycle culminating in disease after some weeks. Vaccination of calves with two doses of 1000 infective larvae attenuated by irradiation is a long-established and effective method of preventing disease. Only healthy calves should be vaccinated and they should be at least 8 weeks old. The vaccine is given 6 and 4 weeks before turnout. Exposure to lightly contaminated pasture will boost immunity, but low-grade patent infections may develop in some animals. Vaccinated and nonvaccinated calves should not be grazed together because the former may contaminate the pasture, enabling lungworm to cycle through the susceptible animals. The vaccine gives a high level of protection under most conditions, but vaccinated calves should not be put onto heavily contaminated pasture. Coughing may occur when immune responses kill lungworm larvae in the lungs. Overt disease can occur in cases of overwhelming challenge. To avoid such problems on severely affected farms, vaccinated calves should be allowed only gradual access to pasture. An experimental recombinant subunit vaccine, based on the parasite’s paramyosin as a recombinant antigen, that overcomes the disadvantages of the attenuated vaccine has been developed.18 In some endemic areas, for example, in the south of Ireland, which has mild winters and an early start to the grazing season, the ideal vaccination program described earlier may be inconvenient and it is possible, by cautiously avoiding periods when massive pasture contamination is likely to occur, to vaccinate at pasture. Calves are sometimes vaccinated at less than 8 weeks old to allow spring-born calves to graze during late summer and autumn, but optimal protection may not be afforded in all cases. Strategic anthelmintic programs provide an alternative to vaccination. The aim is to suppress the infection initiated by overwintered larvae and thereby prevent subsequent contamination of the pasture. This can be done by application of a suitable intraruminal bolus at or just before spring turnout or by giving two or three doses of an avermectin/ milbemycin during the early grazing season.

Diseases of the Bovine Respiratory Tract

VetBooks.ir

These systems are designed to control parasitic gastroenteritis and lungworm. Clinical field experiments have demonstrated good results with ivermectin, fenbendazole, and oxfendazole boluses and with ivermectin treatments given at 3, 8, and 13 weeks after turnout, or doramectin administered at turnout and again 8 weeks later. Calves may become vulnerable after the period of anthelmintic cover if pasture contamination occurs (e.g., because of fungal spread). An extra anthelmintic treatment may be indicated in regions with a very long grazing season. Calves that are exposed to infection but protected by chemoprophylaxis during their first grazing season generally have substantial resistance to reinfection in their second year. Nevertheless, field experiments have shown that immunity can be compromised to a degree related to the level of protection provided. There is concern that such intensive treatment may provoke anthelmintic resistance, but no resistant strains of D. viviparus have yet been reported. FURTHER READING Panciera RJ, Confer AW. Pathogenesis and pathology of bovine pneumonia. Vet Clin North Am Food A. 2010;26:191. Panuska C. Lungworms of ruminants. Vet Clin North Am Food A. 2006;22:583.

REFERENCES

1. Jackson F, et al. Proc Int Conf World Assoc Adv Vet Parasitol. 2007;226. 2. Krzysiak MK, et al. Bull Vet Inst Pulawy. 2014;58:421. 3. Pyziel AM. Acta Parasitol. 2014;59:122. 4. Schunn A-M, et al. PLoS ONE. 2013;8:e74429. 5. Ploeger HW, Holzhauer M. Vet Parasitol. 2012;185:335. 6. Matthews J. Livestock. 2008;13:23. 7. Wapenaar W, et al. J Am Vet Med Assoc. 2007;231:1715. 8. Holzhauer M, et al. Vet Rec. 2011;169:494. 9. Dank M, et al. J Dairy Sci. 2015;In Press. 10. Von Holtum C, et al. Vet Parasitol. 2008;151:218. 11. Bennema S, et al. Vet Parasitol. 2009;165:51. 12. Ploeger HW, et al. Vet Parasitol. 2014; 199:50. 13. Soll MD, et al. Vet Parasitol. 2013;192:313. 14. Kunkle BN, et al. Vet Parasitol. 2013;192:332. 15. Rehbein S, et al. Vet Parasitol l. 2013;192:338. 16. Rehbein S, et al. Vet Parasitol. 2013;192:321. 17. Rehbein S, et al. Parasitol Res. 2015;114:47. 18. Strube C, et al. Vet Parasitol. 2015;8:119.

ATYPICAL INTERSTITIAL PNEUMONIA OF CATTLE (ACUTE BOVINE RESPIRATORY DISTRESS SYNDROME, ACUTE PULMONARY EMPHYSEMA, AND EDEMA) SYNOPSIS Etiology Uncertain; a number of etiologies such as D,L-tryptophan in forage, inhalation of toxic gases and fumes, hypersensitivity to molds, mycotoxicosis, and plant poisonings or feed supplementation with melengestrol acetate have been discussed.

Epidemiology Occurs primarily in adult cattle moved from dry to lush pasture and incidentally in feedlot cattle. Outbreaks or incidental cases of AIP in adult cattle moved from dry to lush pasture in autumn. In feedlot cattle incidental cases are observed toward the end of the finishing period. Signs Outbreaks of acute respiratory distress in pasture form of disease within days of moving to lush pastures; severe dyspnea, open-mouth breathing with extended head and neck, expiratory grunt, subcutaneous emphysema, and rapid death. Subacute form less severe and may survive but develop cor pulmonale later. Clinical pathology None clinically applicable. Lesions Enlarged firm lungs that do not collapse, diffuse congestion and edema, interstitial and bullous emphysema, cranioventral consolidation, hyaline membrane formation, alveolar epithelial hyperplasia, fibrosis. Diagnostic confirmation Lesions at necropsy. Treatment Symptomatic, no effective treatment available. Control Grazing management. Use of antimicrobials to control conversion of tryptophan to 3-methylindole in pastured animals.

Atypical interstitial pneumonia (AIP) of cattle has been known for many years under many different terms, including acute interstitial pneumonia, acute pulmonary emphysema and edema (APEE), acute bovine respiratory distress syndrome (ABRDS), bovine pulmonary emphysema, pulmonary adenomatosis, bovine asthma, pneumoconiosis, and “fog fever.” The syndrome that is characterized by diffuse or patchy damage to alveolar septa is known to occur in an acute and chronic form. The acute presentation, frequently occurring as an outbreak in adult pastured cattle a few days after they are moved from heavily grazed summer pastures to lush fall pastures, is also referred to as “fog fever.” A similarly acute to peracute syndrome in feedlot cattle, primarily affecting animals toward the end of the finishing period, is known as acute or atypical interstitial pneumonia of feedlot cattle. A more chronic form occurring sporadically, often with secondary bacterial involvement, has also been described. The term atypical interstitial pneumonia refers to some clinical characteristics of the syndrome that set it apart from the common acute infectious respiratory tract diseases, especially the viral diseases also causing interstitial pneumonia. Clinically the syndrome is atypical, especially compared with the bacterial pneumonias: • Presentation can be acute or chronic. • Acute or chronic respiratory distress in absence of toxemia

965

• Syndrome is progressive and generally nonresponsive to treatment. • Pathology consists of varying degrees of pulmonary emphysema, edema, hyaline membrane formation, and alveolar epithelial cell and interstitial tissue hyperplasia.

ETIOLOGY The precise etiology of the condition is currently not entirely understood, but because of the obvious epidemiologic differences between the conditions occurring in pastured, housed, and feedlot cattle, it is assumed the AIP can have several different etiologies, all leading to the characteristic lung lesions. The etiologies presented in the following subsections have been proposed. Ingestion of Excessive Amounts of D,L-Tryptophan With the Forage Clinical cases of AIP are frequently reported in adult cattle that have been moved from a dry to a lush pasture in the autumn season. Specific forages have not been implicated, but affected cattle have often been consuming alfalfa, kale, rape, turnip tops, rapidly growing pasture grass, and several other feeds. The levels of tryptophan in lush pasture are sufficient to yield toxic doses of 3-methylindole, the product of tryptophan fermentation in the rumen. A 450-kg cow eating grass at an equivalent DM intake of 3.5% of BW/day with a tryptophan concentration of 0.3% of DM would ingest 0.11 g tryptophan/kg BW/day. The total amount ingested over a 3-day period would approximate the single oral dose of 0.35 g/kg BW needed to reproduce the disease experimentally. However, pasture levels of tryptophan are not necessarily higher in those associated with the disease compared with normal pastures. d,l-tryptophan is converted in the rumen to 3-methylindole (3mI), which, when given orally or intravenously, also produces the lesions characteristic for AIP in cattle and goats. In some naturally occurring cases of AIP in beef cows changed from a dry summer range to a lush green pasture, there is a marked increase in the ruminal levels of 3mI, whereas in other cases the levels are not abnormal. Failure to detect abnormally high levels in the rumen and plasma of naturally occurring cases may be related to the rapid metabolism and elimination of 3mI. Ingestion of d,l-tryptophan has generally been discounted as a possible cause for AIP in feedlot cattle because of its sporadic occurrence and the lack of an epidemiologic association between occurrence of the disease and ration changes. Nevertheless, significantly higher concentration of a 3mI metabolite in the blood of animals affected by AIP compared with healthy control animals were measured in one study, suggesting a possible etiologic role of d,ltryptophan in AIP in feedlot cattle.1

966

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Hypersensitivity to Molds AIP has also been associated with chronic hypersensitivity to moldy hay based on the presence of serum precipitins of the thermophilic antigens of Thermopolyspora polyspora, Micropolyspora faeni, and Thermoactinomyces vulgaris in cattle with allergic alveolitis, a condition also termed as “bovine farmer’s lung.” In Switzerland, a high incidence of serum precipitins against Micropolyspora faeni (60%) and moldy hay antigen (80%) was demonstrated in exposed but apparently healthy cattle from an area where the chronic presentation of bovine farmer’s lung is common. Outbreaks of acute respiratory disease in adult cattle as a result of acute allergic pneumonitis can occur 15 hours after the introduction of severely moldy hay. Serologic investigation and provocative challenge may reveal a hypersensitivity pneumonitis attributable to allergens of Micropolyspora faeni. A hypersensitivity pneumonitis has been produced experimentally in calves by exposure to aerosols of Micropolyspora faeni with or without prior sensitization by subcutaneous injection of the antigen. Although clinically allergic pneumonitis and AIP share a very similar presentation, there are significant pathologic differences indicating that allergic pneumonitis and AIP are different conditions.2 Although hyaline membrane formation, which is characteristic for AIP, is rarely seen with allergic pneumonitis, the latter is typically associated with microscopic granuloma formation that is not seen with AIP.2 Inhalation of Toxic Gases and Fumes Incidental cases of AIP have been reported in cattle exposed to different noxious gases and fumes, such as silo gas, nitrogen dioxide, chlorine gas, or zinc oxide fumes.3 The experimental inhalation of nitrogen dioxide gas is capable of causing acute interstitial pneumonia in cattle and severe alveolar edema and emphysema in pigs, but it seems unlikely that animals of either species would be exposed naturally to a significant concentration of the gas for a sufficiently long period to produce such lesions. Pigs that survived experimental exposure to silo gas did not have the lesions seen in silo-fillers’ disease in humans, and experimental exposure of cattle to nitrogen dioxide gas produces lesions that do not occur in naturally occurring AIP. Acute pulmonary emphysema and deaths have occurred in cattle exposed to zinc oxide fumes produced by the welding of galvanized metal in an enclosed barn housing cattle. Parasitic Infestation For many years it was thought that massive infestation of the lungs by large numbers of lungworm larvae in a lungworm-sensitized animal could cause an allergic reaction resulting in the development of AIP. The

possibility of such hypersensitivity as being associated cannot be totally ignored, but at the present time there is no evidence to support such a theory. Such hypersensitivity may occur when the level of larval infestation of pasture is extremely high, but it is not involved in the great majority of cases. In most cases of naturally occurring AIP, there is no laboratory evidence of lungworm infestation of affected and in-contact animals. Reinfection of cattle with lungworm will occur 2 to 3 weeks following introduction to an infected pasture and cause acute respiratory distress that may be indistinguishable clinically from AIP. The migration of abnormal parasites, particularly Ascaris suis, has been observed to cause an acute interstitial pneumonia in cattle that were allowed access to areas previously occupied by swine. Mycotoxicosis and Plant Poisonings The ingestion of sweet potatoes infested with the mold Fusarium solani has been incriminated as a cause of AIP in cattle. Growth of the mold on the potatoes produces the toxins ipomeamarone and ipomeamaronol and a lung edema factor. The latter is a collective term for a group of substances capable of causing death associated with severe edema and a proliferative alveolitis of the lungs of laboratory animals. It produces a respiratory syndrome that is clinically and pathologically indistinguishable from AIP. The fungus Fusarium semitectum growing on moldy garden beans, Phaseolus vulgaris, which were discarded on pasture, was associated with acute pulmonary emphysema in cattle that consumed the beans and their vines. The fungus produces a pulmonary toxin. The pulmonary toxin 4-ipomeanol (ipomeanol) accumulates in mold-damaged sweet potatoes and induces pulmonary edema, bronchiolar necrosis, and interstitial pneumonia in many mammalian species. Outbreaks have occurred in lactating cows following ingestion of sweet potatoes damaged by Myzus tersicae. Other Fursarium spp. have been found in peanut-vine hay, which has been associated with acute respiratory distress and atypical interstitial pneumonia in adult beef cattle. The population mortality rate as a result of respiratory disease was about 12% and the case-fatality rate 77%. Clinical signs occurred within a few days to 2 months after the animals were fed the peanut-vine hay. A weed, Perilla frutescens, is considered to be a cause of the disease in cattle in the United States and wherever the plant is found. High morbidity and high case-fatality rates are characteristic, and the plant contains a perilla ketone that can be used to produce the disease experimentally. Turf-quality perennial ryegrass straw (Lolium perenne) infected with the endophyte (Acremonium lolii), which yields toxic substances, including lolitrem-B, has been

associated with atypical pneumonia in weaned beef calves. However, feeding the suspect hay resulted in typical ryegrass staggers but not atypical interstitial pneumonia. Melengestrol Acetate Melengestrol acetate (MGA), a feed additive commonly fed to feedlot heifers to suppress estrus, has been associated with AIP based on epidemiologic evidence. Data obtained from Canadian feedlots indicated that the great majority of cases of AIP occurred in heifers and that discontinuing MGA treatment resulted in a reduced number of emergency slaughters, most of which were attributable to AIP.2 Further work did not reveal any effect of oral MGA administration on plasma 3mI concentration. If MGA does play a role in the etiology of AIP, the mechanism through which this occurs is not clear. Bacterial and Mycoplasma spp. Infections There is no evidence that any of the common bacterial pathogens of cattle such as Mannheimia haemolytica, Pasteurella multocida, Histophilus (Haemophilus) somni, or Mycoplasma spp. are primarily associated with AIP. In a series of feedlot cattle with clinical findings consistent with AIP, the pathogens were present in the lung tissues of some animals at necropsy, but their presence was not considered as a primary cause of the pneumonia but rather secondary to the initial injury of the lung that was undetermined. Viral Infections Certain viral infections of the lung may result in interstitial pneumonia. In the interstitial pneumonias caused by the bovine respiratory syncytial virus (BRSV) there is a bronchiolitis and alveolitis, and these should be termed bronchiointerstitial pneumonias. The BRSV is an important cause of outbreaks of acute interstitial pneumonia in beef calves 2 to 4 weeks after weaning. Pathologic evaluation of the lung tissues of feedlot cattle that had acute interstitial pneumonia found that BRSV was not a causative agent. In a series of cases of interstitial pneumonia in feedlot cattle in Saskatchewan, the presence of the BRSV antigen was demonstrated in only 7% of cases, and there was more severe bronchiolar epithelial necrosis than in the other cases that were negative for the virus.

EPIDEMIOLOGY

AIP occurs primarily in adult cows and bulls, usually 4 to 10 days after they are moved abruptly from a dry or overgrazed summer pasture to a lush autumn pasture. The new pasture may or may not have been grazed during that summer, and the species of grass or plants does not seem to make a difference, but usually there is some lush regrowth of

Diseases of the Bovine Respiratory Tract

VetBooks.ir

grass, legume, or other palatable plants. Merely changing pasture fields in the autumn has precipitated the disease. AIP in pastured animals usually occurs in outbreaks, with the morbidity ranging from 10% in some herds up to 50% and higher in others, with a case fatality ranging from 25% to 50%. It is not unusual to observe a mild form of the disease in about one-third of the adults at risk, but only 10% of those at risk may be severely affected. Often, a number of cows are found dead without premonitory signs; many others are severely ill and die within 24 hours. Calves and young growing cattle up to 1 year of age grazing the same pasture are usually unaffected. A retrospective analysis and random sample survey of cattle ranches in northern California found that the type of forage management has a significant effect on the occurrence of the disease. The greatest occurrence of the disease was in herds where the cattle were moved from summer ranges to secondgrowth hay fields or to irrigated pastures or from one irrigated field to another. The adult morbidity rate was 2.6% and the case-fatality rate about 55%. The disease did not occur on ranches with limited or no movement of cattle from summer ranges to lush autumn pastures. The timespan over which the disease occurs during the autumn months is only 2 to 4 weeks. The incidence of the disease declines rapidly following the first frost. The disease has also occurred in the same herd on the same pasture in successive years. AIP in pastured cattle has been recorded in Canada, the United States, Great Britain, Holland, New Zealand, and other countries. The disease is rare in Australia. The disease has been recognized in France for many years as an aftermath disease or aftermath emphysema, especially in the Normandy region. Some reports have suggested a breed predilection, with Herefords being more commonly affected than the Jersey, Holstein, Shorthorn, and Angus breeds, but there are few exact epidemiologic data to support the observation. AIP in feedlot cattle is recognized as important cause of economic loss in feedlot cattle in western Canada and the United States. The disease occurs sporadically, and the incidence is about 2.8% of all cattle placed in feedlots.4 Cases occur most commonly during the summer and fall months, with a higher incidence rate toward the end of the finishing period. In southern Alberta, the disease is most common during hot, dry, and dusty spring and summer days, and typically affects animals expected to be ready for slaughter within 15 to 45 days. Some feedlot operators have observed that the disease is more common in cattle exposed to excessive dust from bedding. In southern Alberta feedlots, the disease occurred late in the finishing period, when animals had been on feed an average of 114 days and weighed 475 kg. All

confirmed cases were in heifers, and plasma concentrations of 3mI metabolites (adducts) were higher in heifers with AIP than in controls. Most of the heifers were receiving melengestrol (MGA) orally to suppress estrus. The odds of an animal with acute interstitial pneumonia being a heifer were 3.1 times greater than the odds that an animal with the disease was a steer. In some large feedlots the estimated relative risk was 4.9. Other types of atypical interstitial pneumonia occur sporadically and may affect only a single animal or several over a period of time. There is not necessarily a seasonal incidence except in areas where cattle are housed and fed dusty and moldy hay during the winter months. AIP has been reported to occur in weaned beef calves about 4 weeks after weaning.

PATHOGENESIS

Because of the number and variety of circumstances in which acute or chronic interstitial pneumonia occurs, it is difficult to suggest a basic underlying cause, or to explain the mechanisms for the development of the lesions and the variations that occur from one circumstance to another. The l-isomer of tryptophan contained in feed is metabolized by ruminal micro­ organisms to indoleacetic acid, which is then converted to 3-methylindole (3mI). The conversion of l-tryptophan to 3mI is maximal at a ruminal pH near neutrality. The 3mI is absorbed from the rumen and metabolized by a mixed-function oxidase system to an active intermediate, which has pneumotoxic properties. Bioactivation of 3mI by alveolar Clara cells leads to profound cellular injury in Clara and type-1 alveolar epithelial cells and, ultimately, atypical interstitial pneumonia. It is postulated that the compound responsible for causing the injury is the electrophilic metabolite of 3mI, 3-methylenedolenine (3mEIN), which forms stable adducts with cellular macromolecules. (Adducts are compounds formed by an addition reaction.) Concentrations of 3mEIN in lung tissue and blood were higher in feedlot cattle that had died of AIP than in healthy feedlot cattle. However, lung tissue concentrations of 3mEIN were similar in samples from cattle with interstitial pneumonia and bronchopneumonia. Mean concentration of 3mEINadduct increased to a maximum value on day 33 and then decreased to a minimum on day 54 after arrival in the feedlot. Plasma 3mI concentrations initially decreased and remained low until after day 54. Neither 3mEIN-adduct concentrations nor plasma 3mI concentrations were associated with deleterious effects on weight gains. The reaction that occurs is a nonspecific but fundamental reaction of the pulmonary parenchyma to a wide variety of insults that may be ingested, inhaled, or produced

967

endogenously. Pulmonary edema is the first morphologic change occurring in ruminants given 3mI. The edema is preceded by degeneration, necrosis, and exfoliation of type I alveolar septal cells. During the acute stage, there is flooding of the alveoli with serofibrinous exudate, congestion, edema of alveolar walls, and hyaline membrane formation. Varying degrees of severity of interstitial emphysema also occur. The interstitial emphysema may spread within the lymphatics to the mediastinum and into the subcutaneous tissues over the withers, over the entire dorsum of the back, and, occasionally, over the entire body, including the legs. If the acute phase is severe enough, there is marked respiratory distress and rapid death from hypoxemia. Unlike the bacterial pneumonias, the emphasis is on edema and proliferation rather than on necrosis. The lesions have been produced experimentally in cattle, sheep, and goats following oral or IV administration of 3mI. Calves appear be more resistant to experimental toxicity with 3mI than adults, which supports the observation that the naturally occurring disease is uncommon in calves grazing the same pasture in which adults are affected. In case the animal survives the acute stage a proliferative stage follows that is marked by proliferation of alveolar type II cells. There is alveolar epithelialization and interstitial fibrosis, the latter being progressive and irreversible. The central features of chronic interstitial pneumonia are intraalveolar accumulation of mononuclear cells, proliferation and persistence of alveolar type 2 cells, and interstitial thickening by accumulation of lymphoid cells and fibrous tissue. Diffuse fibrosing alveolitis is a form of chronic interstitial pneumonia of uncertain etiology, but it is possibly the chronic form of AIP. AIP has been recorded in sheep, and there was extensive alveolar epithelialization. In Norway, an acute respiratory distress syndrome has occurred in lambs moved from mountain pastures onto lush aftermath pasture. The lesions were those of AIP and alveolar epithelial hypersensitivity to molds in the grass is being explored. The experimental oral administration of 3mI to lambs will result in acute dyspnea and lesions similar to those that occur in cattle and adult sheep following dosing with 3mI. However, the lesions in experimental lambs are different from those that occur in lambs affected with the naturally occurring disease.

CLINICAL FINDINGS

This acute form of AIP in pastured cattle is usually obvious. Within 4 to 10 days after adult cattle have been moved onto a new pasture, they may be found dead without any premonitory signs. In the experimental disease, the typical clinical signs of respiratory disease appear within 24 to 36 hours

968

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

after the oral administration of l-tryptophan to adult cattle and within 4 days, 50% of the dosed cows will die. One or several cattle may exhibit labored breathing, often with an expiratory grunt, open-mouthed breathing, head and neck extended, frothing at the mouth, and anxiety. Severely affected cattle do not graze, stand apart from the herd, and are reluctant to walk. If forced to walk, they may fall and die within a few minutes. Moderately affected cattle continue to graze, but their respirations are increased above normal. Coughing is infrequent regardless of the severity. The temperature is normal to slightly elevated (38.5–39.5° C [102–103° F]) but may be up to 41 to 42° C (106–108° F) during very warm weather. There is a similar variation in the heart rate (80-120/min), and those with a rate of more than 120/min are usually in the terminal stages of the disease. Bloat and ruminal atony are common in severe cases. Subcutaneous emphysema is common over the withers and may extend to the axillae and ventral aspects of the thorax. The nostrils are flared, and the nasal discharge is normal. Diarrhea may occur but is mild and transient. Loud, clear breath sounds audible over the ventral aspects of the lung, indicating consolidation without bronchial involvement, are the characteristic findings on auscultation in the early stages of the acute disease. The intensity of the breath sounds may be less than normal over the dorsal parts of the lung if involvement is severe, but in animals that survive for several days the loud crackles characteristic of interstitial emphysema are of diagnostic significance. Most severely affected cases will die within 2 days of onset, but less severe cases will live for several days and then die of diffuse pulmonary involvement. Those that survive longer than 1 week will often have chronic emphysema and remain unthrifty. Of those moderately affected cattle that recover in a few days, some will develop congestive heart failure a few months later, as a result of chronic interstitial pneumonia (cor pulmonale). Calves running with their adult dams will usually not be affected. AIP in nonpastured cattle such as feedlot cattle usually occur sporadically, but several animals may be affected over a period of time. There may or may not be a history of a change of feed or the feeding of moldy or dusty feed. In many cases, a few days will elapse after the appearance of signs before the owner is aware of the affected animals. The animal may have been treated with an antimicrobial for a bacterial pneumonia with little or no response. Dyspnea, increased respiratory effort sometimes with a grunt, deep coughing, a fall in milk production, an absence of toxemia, a variable temperature (38.5-40° C [102-104° F]) and a variable appetite are all common. On auscultation there are loud breath sounds over the ventral aspects of the lungs and crackles over both

dorsal and ventral aspects. The presence of moist crackles suggests secondary bacterial bronchopneumonia. Subcutaneous emphysema is uncommon in these, and most will become progressively worse. Yearling cattle with acute interstitial pneumonia that may be viral in origin may become much worse and die in a few days in spite of therapy. Mature cattle affected with the chronic form of AIP will survive in an unthrifty state with the chronic disease for several weeks and even months. The major clinical features of all these other interstitial pneumonias are obvious respiratory disease, lack of toxemia, poor response to treatment, progressive worsening, and abnormal lung sounds distributed over the entire lung fields. DIFFERENTIAL DIAGNOSIS Atypical interstitial pneumonia (AIP) is usually obvious when presented with an outbreak of acute respiratory disease in adult cattle that have recently been moved onto a new pasture. The onset is sudden; several cattle may have been found dead, and many are dyspneic. Clinical differential diagnoses for AIP include: • Pneumonic pasteurellosis (shipping fever, enzootic pneumonia of calves) that is characterized by fever, toxemia, mucopurulent nasal discharge and less dyspnea; young cattle are more commonly affected, and there is a beneficial response to therapy within 24 hours. • Organophosphatic insecticide poisoning may resemble AIP because of the dyspnea, but additionally there is pupillary constriction, mucoid diarrhea, muscular tremor and stiffness of the limbs, and no abnormal lung sounds. • Nitrate poisoning may occur in cows moved into a new pasture with high levels of nitrate. Many cows are affected quickly, they are weak, stagger, gasp, fall down, and die rapidly. The chocolate brown coloration of the mucous membranes, the lack of abnormal lung sounds, and the response to treatment are more common in nitrate poisoning. • Other interstitial pneumonias in cattle are generally not associated with a change of pasture in the autumn and are difficult to diagnose clinically and pathologically, especially when they occur in a single animal. The chronic and subacute types of interstitial pneumonia are difficult to differentiate from each other and from other pneumonias of cattle. • Extrinsic allergic alveolitis (bovine farmer’s lung) occurs in housed cattle exposed to dusty or moldy feeds for a prolonged period and is characterized by a history of chronic coughing, weight loss, poor milk production, occasionally

green-colored nasal discharge, and dry crackles over most aspects of the lungs. Not infrequently, acute cases occur, and animals die within a week after the onset of signs. • Verminous pneumonia caused by Dyctiocaulus viviparus occurs in young cattle on pasture in the autumn months and causes subacute or acute disease that may resemble AIP clinically but not epidemiologically. Identification of the larvae in the feces or tissues of affected animals should be attempted. • Verminous pneumonia caused by aberrant migration of Ascaris suis larvae may be indistinguishable from acute interstitial pneumonia, but a history of previous occupation of the area by pigs may provide the clue to the diagnosis, which can only be confirmed on histologic examination of tissues.

CLINICAL PATHOLOGY There are no abnormalities of the hemogram or serum biochemistry that have any diagnostic significance. Examination of feces and forage for lungworm larvae will aid in differentiation from verminous pneumonia if past the prepatent period.

NECROPSY FINDINGS

In AIP the lungs are enlarged and firm and do not collapse on cutting. In the early stages of acute cases they contain much fluid that is more viscid than usual edema fluid. The pleura is pale and opaque and appears to be thickened. In peracute cases, the entire lungs are homogeneously affected in this way. Such cases usually have edema of the larynx. In the more common acute case, the lung has a marbled appearance. Adjacent lobes may be affected with any one of four abnormalities. Areas of normal, pink lung are restricted to the dorsal part of the caudal lobes. There are areas of pale tissue indicative of alveolar emphysema, areas of a dark pink color affected by early alveolar exudation, yellow areas in which the alveoli are filled with coagulated protein-rich fluid, and dark red areas where epithelialization has occurred. The latter two lesions are firm on palpation and resemble thymus or pancreas. They are more common in the ventral parts of the cranial lobes. In chronic cases, as a sequela to the acute form described previously, the obvious differences in the age of the lesions suggest that the disease progresses in steps by the periodic involvement of fresh areas of tissue. In all cases there is usually a frothy exudate, sometimes containing flecks of pus, in the bronchi and trachea, and the mucosa of these passages is markedly hyperemic. Histologically, the characteristic findings are an absence of inflammation, except in the case of secondary bacterial invasion, and the presence of an eosinophilic, protein-rich

Diseases of the Ovine and Caprine Respiratory Tract

VetBooks.ir

fluid that coagulates in the alveoli or may subsequently be compressed into a hyaline membrane. This is more apparent in acute cases, and if animals live for a few days, there is evidence of epithelialization of the alveolar walls. In longstanding cases, there is extensive epithelialization and fibrosis. There is a lack of obvious lesions of the small airways, which differentiates interstitial pneumonia from bronchopneumonia. Bacteriologic examination of the lungs is often negative, although in longstanding cases in which secondary bacterial pneumonia has developed, Pasteurella multocida, Mannheimia haemolytica, Streptococcus spp., and Trueperella (formerly Arcanobacterium) pyogenes may be found. A careful search should be made for nematode larvae.

TREATMENT

The treatment of AIP in cattle is empirical and symptomatic because there is no specific therapy available. The lesion is irreversible in severe cases, and treatment is unlikely to be effective. When outbreaks of the disease occur on pasture, the first reaction is to remove the entire herd from the pasture to avoid the development of new cases. However, almost all new cases will usually occur by day 4 after the onset of the outbreak, and removal from the pasture usually will not prevent new cases. Conversely, leaving the herd on pasture usually will not result in additional cases. Severely affected cattle should be removed from the pasture with extreme care, very slowly, and only if necessary, and they should be moved to shelter from the sun. Immediate slaughter for salvage may be indicated in severe cases. Mild or moderately affected cases will commonly recover spontaneously without any treatment if left alone and not stressed, a fact that has not been given due consideration when claims are made for the use of certain drugs. Several different drugs have been advocated and used routinely for the treatment of AIP in cattle. However, none has been properly evaluated, and definitive recommendations cannot be made. Treatment of the chronic interstitial pneumonias is unsatisfactory because the lesion is progressive and irreversible. TREATMENT AND CONTROL Treatment No specific treatment is available for AIP. Control AIP in pastured cattle

Monensin (200 mg/head PO q24h from 1 day before pasture change for at least 4 days after moving to fall pasture) (R-2) Chlortetracycline (2.5 g/head PO q24h from 1 day before pasture change for at least 4 days after moving to fall pasture) (R-2) Lasalocid (200 mg/head PO q24 from 1 day before pasture change for at least 14 days)

CONTROL There are no known reliable methods for the prevention of AIP in pastured cattle, but there are some strategies that merit consideration. Grazing Management If lush autumn pasture contains toxic levels of the substance that causes the acute disease, it would seem rational to control the introduction of cattle to the new pasture. This can be done by controlling the total grazing time during the first 10 days: allow the cattle to graze for 2 hours on the first day, increasing by increments of 1 hour per day, and leave them on full time at the end of 10 to 12 days. If possible, this may be accomplished by rotating cattle back and forth, either between the summer and fall pastures or between the fall pasture and a drylot where ample supply of dry, mature hay is available. Dry, mature hay may be offered ad libitum to adult cattle in the morning before going on pasture for at least 4 days into the grazing period to reduce consumption of pasture forages. Such a management procedure is laborious and may not be practical depending on the size and terrain of the pasture and the holding yards that are available. Inhibition of 3-Methylindole Production in Rumen Controlling the conversion of d,ltryptophan in forage to 3mI is a plausible control strategy. Experimental tryptophaninduced AIP can be prevented by oral administration of chlortetracycline or polyether antibiotics such as monensin. The daily oral administration of 2.5 g/head of chlortetracycline beginning 1 day before and for 4 days following administration of a toxin of l-tryptophan will prevent clinical signs. The daily oral administration of monensin at the rate of 200 mg/head/day beginning 1 day before and for 7 days after an abrupt change from a poor-quality hay diet to a lush pasture reduced the formation of 3mI during the 7 days of treatment, but the effect of the drug was diminished on day 10, 3 days after its withdrawal. Because the effects of monensin on ruminal 3mI are diminished within 48 hours after withdrawal of the drug, effective prevention of acute pulmonary edema and emphysema may require continuous administration of monensin for the critical period of approximately 10 days after the mature animals are exposed to the lush pasture. The daily feeding of monensin in either an energy or protein supplement will effectively reduce ruminal 3mI formation in pasture-fed cattle. Lasalocid at a dose of 200 mg per head once daily in ground grain for 12 days reduced the conversion of tryptophan to 3mI and prevented pulmonary edema. Any combination of these management practices may reduce 3mI production to a greater extent than just providing monensin

969

or implementing grazing management techniques alone. Other Forms of AIP The control of nonpasture cases of AIP depends on the suspected cause and removal of it from the environment of the animals. Every attempt must be made to control the concentration of dust and moldy foods to which cattle are exposed. Feed supplies must be harvested, handled, and stored with attention to minimizing dust and molds. In the preparation of mixed ground feed for cattle, the fineness of grind must be controlled to avoid dusty feed particles that may be inhaled. Because of the creation of dust, the grinding and mixing of dry feeds such as hay, straw, and grains should not be done in the same enclosed area in which cattle are housed. If dusty feeds must be used, they should be wetted to assist in dust control. FURTHER READING Doster AR. Bovine atypical interstitial pneumonia. Vet Clin North Am Food A. 2010;26:395-407. Panciera RJ, Confer AW. Pathogenesis and pathology of bovine pneumonia. Vet Clin North Am Food A. 2010;26:191-214. Woolums AR, McAllister TA, Lonergan GH, et al. Etiology of acute interstitial pneumonia in feedlot cattle: noninfectious causes. Comp Cont Ed Pract Vet. 2001;9:S86-S93.

REFERENCES

1. Loneragan GH, et al. Am J Vet Res. 2001;62:1525-1530. 2. Woolums A, et al. Comp Cont Ed Pract Vet. 2001;9:S86-S93. 3. Doster AR. Vet Clin North Am Food A. 2010;26:395-407. 4. USDA Feedlot 2011, part IV. (Accessed 15.09.15, at: .).

Diseases of the Ovine and Caprine Respiratory Tract ENZOOTIC NASAL ADENOCARCINOMA OF SHEEP AND GOATS (ENZOOTIC NASAL TUMOR) Intranasal adenocarcinoma has been recorded as a sporadic disease of sheep and goats for many years and is now recognized as a contagious neoplasm in these species.1 The disease in sheep and goats is associated with related but different retroviruses, the ovine nasal adenocarcinoma virus (ENT-1) and the caprine adenocarcinoma virus (ENT-2), respectively. These retroviruses are highly conserved, with North American and European isolates being 96% homologous.2 They are homologous with the retrovirus that causes jaagsiekte (JSVR) but can be distinguished by unique sequences of the genome. Nasal adenocarcinoma is not a

Chapter 12 

970

■ 

Diseases of the Respiratory System

VetBooks.ir

component of the disease jaagsiekte, nor are pulmonary tumors present in sheep and goats with nasal adenocarcinoma. Infections with the viruses of enzootic nasal adenocarcinoma and jaagsiekte can occur in the same sheep, and this can potentiate the proliferation of jaagsiekte virus in the infected sheep.

Enzootic nasal adenocarcinoma is recorded in the United States, Canada, Europe, Japan, India, China, and Africa. It is believed to occur on all continents except Australia and New Zealand, but it is not present in the United Kingdom. The disease occurs sporadically but is often clustered in certain flocks and herds, and it is assumed to

A

transmit by the respiratory route. The prevalence in affected flocks varies in different countries. It is generally less than 2% but can be as high as 10% to 15%. There is no seasonal occurrence and no apparent breed or genetic predisposition. There is no apparent influence of nasal myiasis on the prevalence of nasal adenocarcinoma in infected flocks. Clinical disease is recorded occurring as early as 7 months of age, but most occurs in mature sheep between 2 and 4 years of age. Affected animals are afebrile, have a profuse seromucous or seropurulent nasal discharge, and sneeze and shake their heads frequently. There is depilation around the nostrils. The tumor may be unilateral or bilateral. As the disease progresses, there is dyspnea, stertorous breathing with flaring of the nostrils at rest, and open-mouthed breathing following exercise. Some animals develop facial deformity and protrusion of one or both eyes from tumor growth, and the tumor may protrude from the nostril (Fig. 12-17). There is progressive loss of weight, emaciation, and death after a clinical course of 3 to 6 months. There is no detectable immune response in affected animals. At postmortem, the tumor masses are in the ethmoid turbinates, with metastasis to regional lymph nodes in some cases. The tumors may be unilateral or bilateral and are gray or pink in color with a granular surface. The tumors originate in the serous glands of the turbinates and have the histologic features of adenocarcinoma. The disease has been transmitted experimentally in both goats and sheep, with challenge of young kids resulting in disease at 12 to 16 months of age. REFERENCES 1. Radostits O, et al. Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1368. 2. Walsh SR, et al. Virus Res. 2010;151:74.

CONTAGIOUS CAPRINE PLEUROPNEUMONIA SYNOPSIS Etiology Mycoplasma capricolum subsp. capripneumoniae. Epidemiology Highly contagious disease of goats, outbreaks in wild small ruminants do occur Clinical findings Pleuropneumonia. Lesions Pleuropneumonia with no enlargement of the interlobular septa.

B Fig. 12-17  A, Nasal adenocarcinoma in a Suffolk ewe. Notice the seromucous to seropurulent nasal discharge (left greater than right). Very little air movement was detected from the right nostril. B, Endoscopic view of a nasal adenocarcinoma (dorsal pink gray spherical mass) in a Suffolk ram with clinical signs of an upper respiratory tract obstruction.

Diagnostic confirmation Culture, polymerase chain reaction (PCR) on pleural fluid. Latex agglutination test. Treatment Antimicrobials. Control Herd biosecurity; vaccination provides strong immunity but of short duration.

Diseases of the Ovine and Caprine Respiratory Tract

971

VetBooks.ir

Table 12-9  Summary of systemic mycoplasmoses of sheep and goats Bacterial species

Animals affected

Diseases caused

Pathogenicity

M. agalactiae

Sheep/goats

Contagious agalactia, arthritis, pneumonia, granular vaginitis, pinkeye

High

M. arginini

Sheep/goats

Pneumonia, arthritis, vaginitis, pinkeye, mastitis

Low

M. capricolum subsp. capricolum

Sheep/goats

Mastitis and agalactia, pneumonia, arthritis

High

M. mycoides subsp. capri (formerly M. mycoides subsp. mycoides)

Goats

Contagious agalactia, pneumonia, arthritis, high mortality in young kids

Moderate

M. ovipneumoniae

Sheep/goats

Pneumonia

Commonly precursor to pneumonic pasteurellosis

M. putrefaciens

Goats

Mastitis and arthritis

High

Ureaplasma sp.

Goats

Vaginitis

Low

Mycoplasma capricolum subsp. capripneumoniae (formerly strain F38)

Sheep/goats

Contagious caprine pleuropneumonia

High

ETIOLOGY Contagious caprine pleuropneumonia (CCPP) is a classical disease of goats, associated with Mycoplasma capricolum subsp. capripneumoniae and commonly confused with other serious pneumonias of goats and sheep (first isolated in 1976 and previously known as mycoplasma strain F38) (Tables 12-6 and 12-9). The disease was first described in Algeria in 1873 and presents primarily as a pleuropneumonia. The organism is difficult to grow, which can lead to poor differentiation of the CCPP from pneumonic disease induced by other mycoplasmas (M. capricolum subsp. capricolum and M. mycoides subsp. capri). Highly specific PCR tests can differentiate infections within the M. mycoides cluster of goats and will provide more information on the distribution and epidemiology of these diseases.

EPIDEMIOLOGY Occurrence CCPP is one of the most serious fatal diseases of goats in Africa and Asia and has serious socioeconomic effects for sub­ sistence goat herders. It is known as Abu Nini in the Sudan. The exact distribution is uncertain, but clinical disease has been reported from 38 countries, mostly from Africa and Asia, with recent detections in Mauritius (2009), Turkey (2009), and China (2012).1,2 However, the causative organism has only been isolated in some of these countries because of the difficulty growing it and lack of mycoplasmal laboratories. CCPP has many similarities clinically and at necropsy to contagious bovine pleuropneumonia, caused by M. mycoides subsp. mycoides SC, but it is not transmissible to cattle. Sheep can be infected experimentally and sero­ convert, and they have been reported with respiratory disease in Eritrea. Captive and free-ranging wild ungulates, including deer, gazelles, and ibex, can also become infected and suffer disease.1,2 M. capricolum subsp.

capripneumoniae is highly infectious. In newly affected flocks the illness is acute and severe following a brief incubation period (generally 6-10 days, but up to 28 days), with morbidity rates of 90% and case mortality rates of 60% to 100%. The disease is less severe and more sporadic in endemically exposed flocks. Transmission The disease is readily transmitted by inhalation, but the organism does not survive for long in the environment. Infection is brought into the flock by asymptomatic carrier or clinically affected animals. Agent Mycoplasmas are among the fastest evolving bacteria, with high mutation rates.3 Mycoplasma capricolum subsp. capripneumoniae shows a degree of heterogeneity not found among other members of the M. mycoides cluster. Based on sequencing of several genes, including 16SrRNA and H2 locus and other proteins, 24 haplotypes were identified in 25 strains of Mycoplasma capricolum subsp. capripneumoniae and placed within six genotyping groups (A to F), with two distinct evolutionary lineages identified.3 Lineage 1 contains two groups with strains from East Africa, Qatar, Niger, and Mauritius; lineage 2 is subdivided into three groups with strains from the United Arab Emirates, China, and Tajikistan.

CLINICAL FINDINGS

All ages and sexes are affected. Clinical findings are restricted to the respiratory system and include cough, dyspnea with an extended neck, painful cough, and fever (40.5–41.5° C [104.5–106° F]). Animals often lay down, although they can stand and walk, and continue to eat and ruminate. In the terminal stages there is rapid respiratory rate, mouth breathing, tongue protrusion, and frothy salivation, followed by death in 2 or more days. In less acute infections the clinical signs

are milder, and coughing may only occur following exercise. Under adverse climatic conditions or in kids the disease may occur in a septicemic form, with little clinical or postmortem evidence of pneumonia. Outbreaks of CCPP in gazelle in the Middle East had similar clinical and pathologic signs to goats, although there was often sudden death.2

CLINICAL PATHOLOGY

Antigen can be detected in lung tissue and pleural fluid by PCR based upon the 16S rRNA genes. A real-time PCR offers advantages over conventional PCR including speed, greater specificity and sensitivity, and elimination of post-PCR processing.4 Serologic tests used to identify carrier animals include complement fixation, ELISA, and a latex agglutination test. The latter is robust, available commercially, and suitable for field use. Monoclonal antibody is used in serologic tests to identify caprine isolates by the disc growth inhibition method, which will include M. agalactiae, M. capricolum subsp. capricolum, and the other members of the M. mycoides cluster associated with goats. A competitive ELISA using monoclonal antibodies is highly specific for CCPP.5

NECROPSY FINDINGS

The necropsy findings are similar to those of contagious bovine pleuropneumonia except that there is no thickening of the interlobular septa. Lesions are restricted to the lungs (often one lung) and pleura, with hepatization, increased pleural fluid, and a fibrinous pleuritis, which differentiates the disease from that caused by M. mycoides subsp. capri. Histologically, there is acute serofibrinous to chronic fibrino-nectoric pneumonia with interstitial intralobular edema, rather than a thickening of the interlobular septa that occurs with other mycoplasmal infections. Inflammatory exudate consists mainly of neutrophils. There is also peribronchiolar lymphoid hyperplasia.

972

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Samples for Confirmation of Diagnosis • Bacteriology—Pleural fluid and lung from the interface of the hepatized and normal lung tissue. These mycoplasmas are fragile and should be freeze dried or placed in transport medium if there is to be a significant transport time (>2 days). Conventional or real-time PCR can be performed on samples of pleural fluid dried on filter paper. • Serology—CFT, Latex agglutination, competitive ELISA5 DIFFERENTIAL DIAGNOSIS The other pulmonary mycoplasmoses from which this disease needs to be differentiated are those associated with Mycoplasma mycoides subsp. capri (formerly M. mycoides large colony type) and M. capricolum subsp. capricolum.

TREATMENT Clinical cases respond to a range of antibiotics, including intramuscular tylosin (10 mg/ kg BW), oxytetracycline (15 mg/kg/d), or tilmicosin, marbofloxacin, and danofloxacin.6-8 The severity of the disease is reduced, but treated animals are still sources of infection.

CONTROL

Effective biosecurity measures are needed to prevent the introduction of the disease into a flock via contact with infected carriers. Killed vaccines effectively reduce morbidity and mortality rates. These have been widely used in many countries, although they can be of variable quality.2 Immunity is generally short lived, and so a booster 1 month after the first vaccination provides additional protection. There is little evidence that maternal antibody interferes with the development of immunity, but kids born to does that have been vaccinated while pregnant are often not vaccinated before 10 to 12 weeks of age. Live attenuated vaccines have been trialed but are not yet commercially available and may not be permitted in some jurisdictions.9 FURTHER READING Nicolas R, Churchward C. Contagious caprine pleuropneumonia: new aspects of an old disease. Transbound Emerg Dis. 2012;59:189-196. Prats-van der Ham M, et al. Contagious caprine pleuropneumonia (CCPP) and other emergent mycoplasmal diseases affecting small ruminants in arid lands. J Arid Environments. 2015;119:9-15. Radostits O, et al. Contagious caprine pleuropneumonia. In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1140-1141.

REFERENCES

1. Prats-ven der Ham M, et al. J Arid Environ. 2015;115:9.

2. Nicholas R, Churchward C. 2012;59:189. 3. Depuy V, et al. Vet Res. 2015;46:74. 4. Fitzmaurice J, et al. NZ Vet J. 2008;56:40. 5. Peyraud A, et al. BMC Vet Res. 2014;10:48. 6. Ozdemir U, et al. Trop Anim Health Prod. 2006;156:286. 7. Srivastava AK, et al. Vet Rec. 2010;167:304. 8. Balicki E, et al. Small Rum Res. 2008;77:75. 9. Tarekegn S, et al. Afr J Microbiol Res. 2012;6:3085.

nominated as causes. M. haemolytica is a common secondary infection and may lead to more acute, suppurative respiratory disease. The disease, which might be most accurately identified as chronic undifferentiated enzootic pneumonia of sheep, is probably a collection of etiologically specific diseases.

CHRONIC ENZOOTIC PNEUMONIA OF SHEEP (CHRONIC NONPROGRESSIVE ATYPICAL PNEUMONIA, SUMMER PNEUMONIA, PROLIFERATIVE EXUDATIVE PNEUMONIA)

M. ovipneumoniae M. ovipneumoniae is now considered to be one of the more important parts of the disease complex, and may be the initiating cause.1 It is commonly isolated in large numbers from the lungs of affected sheep, but it can also be isolated from the nasal cavity of some normal sheep and less occasionally from normal lung.1 Experimental challenge with pure cultures of the organism produces minimal lesions, but aerosol or intrabronchial challenge with homogenates of affected lung that contain the organism produces proliferative interstitial and lymphoid pneumonic lesions indistinguishable from the natural disease. M. ovipneumoniae is a facultative pathogen that requires compromised lung defense mechanisms to initiate lesions; infection with this organism subsequently predisposes the lung to secondary infection with organisms such as Past. haemolytica. There is considerable heterogeneity in M. ovipneumoniae, and several different strains may be isolated from a pneumonic lung.2 Differences between strains in pathogenicity are not determined. Other mycoplasma, including M. mycoides subsp. mycoides, M. mycoides subsp. capri, M. putrifasciens, and M. argininii, may be associated with chronic enzootic pneumonia in tropical zones, but M. argininii is considered to have no role in atypical pneumonia of lambs in the United Kingdom.3

SYNOPSIS Etiology Multifactorial, with Mycoplasma ovipneumoniae, viruses and secondary bacterial infections implicated. Epidemiology Affects sheep under 12 months of age. Seasonal occurrence, summer and autumn in southern hemisphere. Common disease affecting most flocks, but severity varies between farms. Clinical findings Insidious onset. Coughing, nasal discharge and uneven weight gain in a mob. Lesions Consolidation of anterioventral lobes of lung. Pleuritis. Diagnostic confirmation Postmortem lesions. Treatment Antimicrobials for severely affected individual sheep. Control No effective control procedure established.

Enzootic pneumonia is defined here as the common, lowly pathogenic disease of sheep, particularly lambs, which is common in all sheep populations. The disease is recognized by different names in different areas of the world. It can be differentiated from the acute fibrinous pneumonia and pleurisy associated with Mannheimia (Pasteurella) haemolytica, which is often called enzootic pneumonia in the British literature, and from the chronic progressive pneumonias, maedi and jaagsiekte.

B. parapertussis B. parapertussis is a common isolate from the nasal cavities and lungs of sheep with chronic enzootic pneumonia in New Zealand and is also believed to have an initiating role in the disease. It produces a cytotoxin that damages ciliated epithelium in the trachea and experimental challenge of colostrum-deprived lambs produces mild pulmonary lesions similar to those seen early in the natural disease. B. parapertussis also can predispose pneumonic pasteurellosis.

ETIOLOGY

Parainfluenza-3 (PI-3) Virus PI-3 is a cause of a mild undifferentiated pneumonia in sheep, and surveys around the world have shown that it is a widespread infection. The disease is clinically mild and marked by the presence of interstitial pneumonia. Antibodies to PI-3 are present in lambs soon after birth, but the half-life is short, and lambs are susceptible by the time they are weaned and mixed with other lambs, which is when clinical disease often occurs. In the experimentally produced

Although the disease is well known, its cause is not well defined. This is partly because of its nonfatal character, which leads to incomplete examination of early cases; most of those submitted for examination or necropsy are distorted by secondary bacterial infections. It has a multifactorial etiology, with a mix of (mainly) Mycoplasma ovipneumoniae, Bordetella parapertussis, chlamydia, parainfluenza-3 (PI-3) virus, adenovirus, a respiratory syncytial virus, and reovirus

Diseases of the Ovine and Caprine Respiratory Tract

VetBooks.ir

disease in lambs there is a slight seromucosal nasal discharge, coughing, increased sensitivity to tracheal compression, and fever of 40° to 41° C (104–106° F). At necropsy there is obvious hyperemia of the upper respiratory mucosa, including the trachea; the bronchial lymph nodes are enlarged; and there are small foci of catarrhal inflammation of pulmonary parenchyma of the apical and cardiac lobes. However, challenge of lambs at 2 weeks of age with this virus and M. haemolytica, although producing disease, did not result in prolonged disease lasting to slaughter, and it was concluded that these agents, without other factors, were not the cause of enzootic pneumonia. This conclusion is supported by the results of vaccine trials with PI-3 against enzootic pneumonia.4 Bovine Respiratory Syncytial Virus BRSV has resulted in pneumonia following experimental challenge of sheep and is evidenced clinically by fever and hyperpnea and pathologically by multifocal pulmonary consolidation and necrosis of epithelial cells. There is little evidence for BRSV as a cause of significant respiratory disease in sheep. Other Agents Adenovirus and a type-3 reovirus have been used experimentally to produce pneumonic lesions, and a vaccine has been produced to protect lambs against the adenovirus infection. Similarly, sheep herpesvirus, caprine herpesvirus-1, will produce an interstitial pneumonia in experimentally challenged SPF lambs, but there is no evidence of a causal association with chronic enzootic pneumonia. Autoantibodies to upper respiratory cilia have been detected in sheep colonized with M. ovipneumoniae, and it is suggested that they contribute to the pathogenesis of coughing in this disease.

EPIDEMIOLOGY Occurrence Enzootic pneumonia affects animals up to 12 months but may commence as early as 6 weeks of age. The disease can occur in both lambs at pasture and housed lambs. In many affected flocks, 80% of 4- to 5-month-old lambs have clinical signs and lesions, and the disease is credited with causing a significant depression in growth rate after weaning in lamb flocks with a high prevalence. This has been confirmed in controlled studies on the effect of the experimentally produced disease on weight gain in housed and pasture-fed lambs. Enzootic pneumonia has a seasonal pattern that differs according to locality and management. In Australia and New Zealand, the period of peak prevalence is in the late summer and autumn. In a longitudinal slaughter study of lambs in New Zealand, the

prevalence of pneumonic lesions was found to increase from early summer to early autumn, with an overall prevalence of pneumonia of 42%. There were significant differences in prevalence between different regions of the country. Factors such as comingling sheep from different sources and environmental stress can precipitate clinical disease. Environmental Risk Factors In Australia and New Zealand, clinical outbreaks of enzootic pneumonia in lambs aged 5 to 8 months are often associated with heat stress, yarding after weaning, use of plunge or shower dips, and transport or mustering of sheep in hot dry conditions. Cases commence within 1 to 3 weeks after transport. In contrast, in the United Kingdom and Europe this disease occurs primarily in the late winter and early spring; in the more intensive production systems of the northern hemisphere, the disease is commonly associated with environmental problems of housing. In Ireland, an association has been made between the occurrence of lesions at slaughter and the extent of rain and windchill experienced by the sheep in the 2 months before slaughter. Economic Importance Death loss from this disease is minor, but economic loss is considerable and includes reduced growth rate, prolonged periods on the farm before reaching slaughter weight, the drug and labor costs associated with treatment, slaughterhouse wastage, and downgrading of carcasses with pleural adhesions and an effect on carcass quality. The situation is similar to that with enzootic pneumonia of pigs.

CLINICAL FINDINGS

The disease is insidious in onset and can persist in a group of lambs for 4 to 7 months. The disease has mild clinical manifestations, with the primary signs being poor and uneven weight gains, an increased nasal discharge, coughing, increased respiratory rate, and respiratory distress with exercise. Increased intensity and a higher pitch of breath sounds are heard on auscultation over the region of the bronchial hilus, and sounds of fluid in the airways are heard in some cases at rest but can usually be elicited by inducing the lamb to cough. There may be periods of fever. There is a relationship between the proportion of the lung affected with pneumonia and average daily gain, and in one study weight gain was reduced by 50% when greater than 20% of the lung was affected. The weight loss is most apparent clinically soon after the disease commences.

NECROPSY FINDINGS

At postmortem there are clearly demarcated areas of consolidation in the anterioventral lobes, and there may be pleuritis with pleural

973

adhesions. The diagnosis is on gross lesions and the presence of typical lesions on histologic examination.

TREATMENT AND CONTROL

Treatment is not usually undertaken unless there is secondary infection to produce acute respiratory disease. Nevertheless, lincomycin (5  mg/kg IM), given twice or three times at intervals of 2 days, and oxytetracycline (20  mg/kg, IM), given twice at 4-day interval, both gave good clinical cure and increased growth rates in a study in Greece.5 Control is based on the avoidance of stress factors that can exacerbate existing infection. FURTHER READING Radostits O, et al. Chronic enzootic pneumonia of sheep (chronic nonprogressive pneumonia, summer pneumonia, proliferative exudative pneumonia). In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1361-1362. Scott PR. Treatment and control of respiratory disease in sheep. Vet Clin North Am Food A. 2011;27:175-186.

REFERENCES

1. Sheehan M, et al. The Vet J. 2007;173:630. 2. Parham K. Vet Microbiol. 2006;118:83. 3. Lin Y-C, et al. Res Vet Sci. 2008;84:367. 4. Thonney ML, et al. Small Rum Res. 2008;74:30. 5. Skoufos J, et al. Small Rum Res. 2006;66:214.

OVINE PROGRESSIVE PNEUMONIA (MAEDI, MAEDI-VISNA) Ovine progressive pneumonia and maedi are North American and European terms for slow virus diseases of sheep in which a chronic progressive pneumonia is a major manifestation. The name maedi is derived from the Icelandic term for dyspnea. Maedivisna virus can also produce visna, which is a disease of the nervous system and is discussed elsewhere under that heading. Additional manifestations of infection are arthritis, indurative mastitis, and ill-thrift. These diseases have a close relationship with caprine arthritis encephalitis. La Bouhite and Graff–Reinert disease are local names for maedi in France and South Africa, respectively. In the United States it was originally described as Montana progressive pneumonia, and in Holland as zwoergersiekte. SYNOPSIS Etiology Ovine retroviruses Epidemiology Most sheep infected as lambs. Persistent infection. High prevalence of infection in many countries but low prevalence of clinical disease. Transmission is via infected colostrum and milk, but lateral transmission also occurs.

Continued

974

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Clinical findings Clinical disease of mature sheep, long incubation, long clinical course. Dyspnea and respiratory distress, initially with exercise but eventually also at rest. Some sheep also manifest chronic wasting and/or indurative mastitis. Necropsy findings Lungs uniformly increased in bulk with enlargement of bronchial and mediastinal lymph nodes. Lymphocytic interstitial pneumonia. Discrete or diffuse hardening of mammary glands with lymphoid infiltration. Diagnostic confirmation Clinical signs, pathology and serology. Polymerase chain reaction (PCR) provides confirmation of infection. Treatment None. Control Segregated rearing. Test and cull.

ETIOLOGY Maedi-visna virus (MVV) and ovine progressive pneumonia virus (OPPV) are single-stranded RNA nononcogenic ovine lentiviruses within the retrovirus family. They have a tropism for monocytes, macrophages, and dendritic cells, but not T-lymphocytes. This is an important determinant of their pathogenesis because they induce a persistent infection in sheep that can cause lympho­ proliferative changes in the lung, mammary tissues, brain, and joints. There is a high degree of relatedness with the lentivirus associated with caprine arthritis encephalitis (CAE), and these ovine and caprine lentiviruses share nucleotide homology and serologic properties. Consequently, MVV, OPPV, and CAE viruses are now regarded as a viral continuum and referred to as small ruminant lentiviruses (SRLV).1 Isolates of MVV from naturally infected sheep are genetically heterogeneous, antigenic drift is common, and antigenic variation of the surface protein facilitates the persistence of the virus in the host, both as latent and chronic infections. There is evidence for variation in pathogenic potential between isolates and hosts, and it is estimated that only about 30% of infected animals develop disease.2 There is some evidence that the North American strains of ovine lentivirus may have originated from cross-species transmission of caprine arthritis–encephalitis virus rather than from maedi lentivirus. However, the similarity in clinical manifestation of maedi and ovine progressive pneumonia and the evidence that the causative viruses are part of a continuum permit the discussion of these diseases as a single entity.

EPIDEMIOLOGY Occurrence The earliest reports of the disease were from South Africa and the United States, but it now occurs in all major sheep-producing

countries with the exception of Australia, New Zealand, and Iceland. Maedi virus was present in Iceland, being introduced in 1933 through the importation of infected Karakul sheep, but was eradicated in 1965. Because of the susceptibility of the local sheep and management practices that favored transmission, it developed to a problem of major national significance. In individual flocks the annual mortality was often 15% to 30%, and in these circumstances sheep farming was not economically viable. Approximately 105,000 sheep are believed to have died of the disease, and 650,000 sheep had to be slaughtered to eradicate it from the country. The international movement of sheep has facilitated the spread of the disease, and it is believed to have been introduced into Denmark, Norway, Sweden, and Great Britain since the 1970s through the importation of infected sheep. However, as yet no other country has experienced the severity of disease that occurred in Iceland. Host Range Sheep and goats are the only species known to be susceptible to MVV, and infection cannot be established by experimental challenge in cattle, deer, pigs, dogs, horses, chickens, mice, and rats. Rabbits are susceptible, but infection is limited to the acute stage before the production of antibody; chronic infection, as seen in sheep and goats, does not occur. Hybrid mouflon–domestic sheep and calves have been experimentally infected with CAE virus, although the infection was cleared naturally in the latter, and SRLV nucleic acid detected in wild ibexes in close contact with goats.2 A serologic survey of wildlife in the United States found no evidence of infection in bighorn sheep, elk, white-tail deer, or antelope. All breeds of sheep appear to be susceptible to infection, but there may be differences in breed susceptibility, based on differences in seroprevalence in flocks with more than one breed of sheep. These differences are not consistent and so they may reflect differences in the susceptibility of family lines within a breed. Variations in the gene encoding an ovine transmembrane protein (TMEM 154) are associated with increased or decreased susceptibility of sheep to SRLV; sheep with glutamate at position 35 have increased susceptibility, whereas those with lysine or a deletion have decreased susceptibility.3 The average frequency of highly susceptible alleles across 74 sheep breeds was 0.51, with more than 25% of mainstream breeds being greater than 0.8.4 In contrast, frequency of highly susceptible alleles across 3 hill breeds was 0.26 to 0.42, suggesting that they would be less affected by MVV infection.5 Prevalence The prevalence of infection varies between farms, breeds, and countries. In the United

States, infection is more common in the western and midwestern and uncommon in the southern states. An estimate of flock seroprevalence (flocks with 1 or more positive sheep) of 48%, and overall seroprevalence of 24%, was recorded in samples collected in 2001 from sheep in 29 states. More recently, samples collected in Wyoming in 2011 found a flock prevalence for OPPV of 47.5% and overall seroprevalence of 18%, with open range (unfenced) flocks at significantly higher risk compared those that were fenced in (an odds ratio of 3.5).6 In Canada, a random survey found a flock prevalence of 63%, with 19% of sheep over 1 year of age being seropositive, and more recently a flock prevalence of 25% in Manitoba. Serologic surveys that use the AGID test will markedly underestimate prevalence, demonstrated by a study in Alberta where 27% of culled ewes were positive on histopathology, but using the AGID there was a seroprevalence of only 13%.7 In the United Kingdom, MVV was introduced more recently, and so this country has lower flock and within-flock prevalence rates (estimated at 3% and up to 15%, respectively, in 2010). However, there is concern that the prevalence of maedi-visna may be increasing, demonstrated by a steady increase in the number of introduced infections in approximately 2600 flocks that participate in a maedi-visna accreditation scheme. These breakdowns are usually caused by flocks not adhering to the biosecurity rules of the scheme, such as introducing sheep into a nonaccredited flock on the same holding.8 There is considerable variation in the prevalence of seropositive sheep between flocks. Rates of seropositivity increase with age, and so within-flock seroprevalence is influenced by the average age of the flock. Flock seroprevalence also has been positively associated with the use of foster ewes, allowing lambs older than 1 day to have contact with other lambing ewes, flock size, close contact during confinement for lambing, stocking density on pasture, and the length of time that the flock has been in existence. Within flock seroprevalence is much higher in flocks that are also infected with pulmonary adenomatosis compared with those that are not. Transmission The disease is spread most commonly by inhalation of infected aerosols and the ingestion of infected colostrum or milk. Vertical transmission following in utero infection is possible but relatively uncommon. Virus is also shed in the semen of infected rams if there are leukocytes in the semen, and so this risk may be increased in rams that are also infected with Brucella ovis. Lambs may contract the infection at or shortly after birth, either from contact with infected ewes or from ingestion of infected colostrum and milk. The virus then infects

Diseases of the Ovine and Caprine Respiratory Tract

VetBooks.ir

dendritic cells at the mucosal surface, and these migrate to local lymph nodes, where virus is transferred to macrophages and spreads systemically.2 Alveolar macrophages play a similar role when infection occurs via the respiratory route. Lambs born to seropositive ewes have a significantly greater risk of infection than those from seronegative ewes, and lambs born to ewes that have been infected for a long time are at greater risk of infection. The chance of transmission to lambs from infected ewes increases with the period of contact, but it can occur within the first 10 hours of life. Lateral transmission can occur in older sheep, and this was probably important in the transmission of the disease in Iceland and the Basque area of Spain, and it has been a significant component of the spread of infection of the virus in flocks in the United Kingdom. In some flocks, the spread of infection can be rapid, and the majority of the flock can seroconvert within a few years of the introduction of infected sheep. The spread of infection is often rapid in flocks that are concurrently infected with the retrovirus causing pulmonary adenomatosis. There are many macrophages in the lungs of these sheep, and so these cells will also be infected with MVV if it is present. In dualinfected sheep, the copious lung fluid produced by sheep with pulmonary adenomatosis contains MVV and can increase the risk of lateral transmission of maedi-visna. Economic Importance Economic loss from this disease is associated with increased mortalities, decreased longevity, decreased value of cull animals, and reduced productivity associated with subclinical infections, such as failure to thrive or to rear lambs. Losses are usually more severe in intensive housed operations, and they can be catastrophic in flocks that derive a large proportion of income from the sale of breeding animals (stud or seedstock flocks). Clinical disease occurs in sheep 2 years old or older, usually in sheep 3 to 4 years of age. Severe disease is more likely when the within-flock prevalence exceeds 50% and has a case-fatality rate of 100%.9 Up to 30% of infections are subclinical, and so clinical disease may not be obvious or common in flocks that have a low prevalence. A high proportion of infected sheep, premature culling, and high mortality rates have occurred in flocks in Iceland, the Netherlands (particularly the Texel breed), the United States, the United Kingdom, and other European countries, especially in intensively managed dairy flocks. It is possible that the major economic loss associated with infection with these viruses rests with the effects of subclinical infection on productivity of infected flocks. Subclinical infection of breeding ewes in some flocks has been associated with a reduction in conception rate and lowered birth weights and/

or reduced growth rates in their lambs. The reduction in growth rate of the lamb is associated with indurative mastitis and a lowered milk intake. This may be expressed by a decreased growth rate of lambs from only older ewes. In other flocks there has been no evidence of effect on the birth weight or growth rate of lambs born of infected ewes. Subclinical infection has no effect on mature ewe body weight or greasy fleece weight.

PATHOGENESIS

SRLV infections are classed as immunopathologic diseases, whereby the host immune response is responsible for most of the pathology rather than the virus itself. The virus infects cells of the monocyte/ macrophage lineage and attaches to cells by the binding of its envelope glycoprotein to specific receptors on the cell surface. The virus replicates its RNA genome via a DNA intermediate provirus that is integrated into the chromosomal DNA of infected cells. With initial infection there is virus replication; this is followed by an immune response that restricts viral replication but fails to eliminate the virus completely. The immune response occurs between 2 and 8 weeks after infection, with antibody to different viral antigens emerging at different times during this period, although some sheep do not develop an antibody response until several months after infection. The ability to establish latent infection of monocytes, which then transfer virus to other organs, may be related to the fact that SRLV are relatively poor at inducing type 1 interferon (IFN), an important mediator of immunity to viral infection.2 In monocytes, replication is restricted and does not proceed beyond the synthesis of provirus in most infected cells. The principal site of virus replication is the macrophage, and pulmonary secretions and milk containing infected macrophages are the main source of virus for natural transmission. Diseases such as pulmonary adenomatosis, which increase the number of macrophages in lung secretions, will facilitate transmission of ovine progressive pneumonia virus via aerosols. The replication of virus initiates viralspecific immune responses (immune activation), and immune-mediated lesions develop in various organs. Production of viral antigen attracts more monocytes, which become latently infected, and so a cycle of latent infection and immune activation, with lymphocytic hyperplasia, is established.2 The infected macrophages in the affected tissues are surrounded by a slowly progressing inflammatory response, creating a focus of mononuclear cell aggregation. Many tissues can be involved, but the lungs, mammary gland, central nervous system and joints are most affected. Any or all of these organs can be affected in a single sheep, but genetic differences in host susceptibility and the virus

975

often lead to a predominance of a single syndrome in a flock. For example, Border Leicesters in the United States and Texels in Holland appear more susceptible to lung infection (maedi), whereas Icelandic sheep are more susceptible to the central nervous disease (visna). In the lung there is a gradual development of a lymphocytic interstitial pneumonia dominated by CD8+ T lymphocytes, hyperplasia of smooth muscle, and fibrosis.2 There is no healing or shrinkage of tissue, so the lungs increase in size and weight, alveolar spaces are filled, and dyspnea and anoxia gradually develop. The pathologic lesions develop very slowly during the preclinical and clinical stages of the disease, so that they are widespread and there is little ability to compensate when clinical signs do appear. In the central nervous system there is infiltration of the meninges and white matter with lymphocytes. The demyelination that occurs in visna is believed to result from the direct effect of the virus on oligodendrocytes and astrocytes and is believed to be the result of an inflammatory response provoked by the presence of viral antigen in these cells. Similar infiltrations occur in the udder. Lymphoid follicles are found in the alveolar parenchyma, often with atrophy of the alveolar tissue. Numerous lymphocytic follicles also occur around the lactiferous ducts, some of which may be occluded by lymphocytic aggregates protruding into their lumens.

CLINICAL FINDINGS

There is a long incubation period. Clinical disease, if it occurs, does not develop before 2 years of age, and most clinically affected sheep are older than 3 years. The clinical signs develop insidiously and progress slowly, and there is a long clinical course. The earliest signs are usually listlessness and loss of body condition that progresses to emaciation. The presenting syndrome can be one of an increased cull rate of ewes in poor condition. Signs of respiratory involvement are not evident in the initial stages of the disease, but there is exercise intolerance, and affected sheep will fall back behind the flock when the flock is moved. Dyspnea, with an increase in respiratory rate (80–120/min at rest) and flaring of the nostrils, or openmouth breathing, develops later, but there is no evidence of excess fluid in the lungs. There may be coughing and some nasal discharge, but in most instances this occurs in sheep with secondary bacterial pneumonia. The body temperature is in the high normal range, and there may be inflammation of the third eyelid. Clinical illness lasts for 3 to 10 months, and the disease is always fatal. Clinically affected sheep are more prone to other diseases, such as pregnancy toxemia. In some sheep, clinical respiratory disease is minimal, and the major manifestation is wasting and the thin ewe syndrome.

976

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Indurative mastitis (“hard bag” or “hard udder”) also has an insidious onset, with ewes usually in their third or later lactation before the disease becomes clinically obvious, although histologic changes will be apparent far sooner. In early stages, hardening of the udder is more easily detected at drying off. In advanced cases the udder is enlarged and uniformly very firm, but the teats are limp. There is very little milk in the teat cistern, although it appears normal.9 Mammary involvement may occur, along with signs of respiratory infection, or affected ewes may show no other clinical abnormality. The lambs of ewes with less severe involvement may have a reduced growth rate. Arthritis is occasionally seen in naturally infected sheep, usually when they are from 1 to 6 years of age. These sheep become lame and emaciated, with obvious swelling of the carpal joints.

CLINICAL PATHOLOGY

There is a progressive, moderate hypochromic anemia, with hemoglobin levels falling from 12 to 14 g/dL to 7 to 8 g/dL, and some depression of the red cell count. There is a tendency to leukocytosis, which in experimental infections is quite marked between exposure and the onset of clinical disease, but the count returns to normal when clinical signs appear. There is also hypergammaglobulinemia. There is an increase in the number of lymphocytes and neutrophils in bronchoalveolar lavage fluid, with more CD8+, fewer CD4+, and an inversion of the CD4+/CD8+ cell ratio. In clinical cases, diagnosis is by the presence of the appropriate clinical syndrome, supported by the presence of a positive serologic test for the virus. A positive serologic test, by itself, has limited value for the diagnosis of individual sheep because there is a high prevalence of seropositivity in many flocks, especially in older animals. A positive test indicates that the animal is infected, but does not indicate that signs or lesions are attributable to infection with the virus. Detection of Antigen PCR is a sensitive method for detection of small amounts of viral nucleic acid, but it may not be available for routine diagnosis in some jurisdictions. It has been used to detect antigen in the third eyelid of infected sheep. Serologic Tests Assessing flock status (the presence or absence of infection) and the status of an individual sheep currently relies on serologic testing. The agar gel immunodiffusion (AGID) and ELISA tests are used in most countries. The AGID test is easy to perform and inexpensive, and thus it is often the most commonly used routine diagnostic test. It has a high specificity but often a lower

sensitivity than the indirect and competitive ELISA tests, which may vary depending on the antigen used.10 Sensitivities of these tests vary from 64% to 97%, and thus they will be unsuited for diagnosis of infection in individual animals or use in test and cull programs if at the lower end of this range. The value of serologic testing rests primarily with the establishment of the infection status of the flock. A negative test in an individual sheep could mean that the sheep is free of infection, but this result can also occur in an infected animal that has not yet responded to infection. A commercial indirect ELISA using a recombinant core protein and a synthetic transmembrane protein as antigens was developed in the Netherlands. Although it had high sensitivity and specificity, it was labor intensive and expensive, and so a pooling procedure that required modifications to the test was developed.11 Subsequently, testing of bulk milk tank samples by ELISA or PCR was confirmed as a cost-effective alternative for flock testing and capable of detecting early infection in dairy flocks. The ELISA detected a within-herd prevalence of less than 1% when samples were tested undiluted and less than 3% when using samples diluted 1 in 10.12 All the bulk milk samples from known SRLV-free flocks (138) tested negative, whereas 50% of samples from flocks with an unknown SRLV status (111) were positive.12 Agreement between the ELISA and two real-time PCR tests on a subsample of 59 milk samples was 90% for the LTPCR and 98% for the leadergag PCR.12

NECROPSY FINDINGS

Lesions may be present in the lungs and associated lymph nodes, brain, joints, mammary gland, and blood vessels, but gross lesions in most sheep are confined to the lungs and, in some cases, the mammary glands. In advanced cases, the lungs are larger and 2 to 4 times as heavy as normal lungs. They collapse much less than normal when the chest is opened, and are gray–blue to gray–yellow in color. There is a diffuse thickening of both lungs, with abnormal color and consistency in all lobes and consistent enlargement of the bronchial and mediastinal lymph nodes. Histopathologic changes are characteristic of a chronic interstitial pneumonia, with proliferation of lymphoid tissue and the presence of numerous lymphoid follicles. There is infiltration of lymphocytes and macrophages in the interalveolar septa, which are thickened, and the bulk of the alveolar space is replaced by the thickened alveolar walls. Larger airways are unaffected. There is a complete absence of healing, consistent with the progressive nature of the disease, and vasculitis is often present. Lesions of arthritis, encephalitis, and mastitis are often present. The mastitic lesion

comprises an interstitial accumulation of lymphocytes and the presence of periductal lymphoid nodules with atrophy of alveolar tissue. Culture of the virus is difficult, and confirmation of the diagnosis is often limited to the presence of characteristic microscopic lesions, preferably supported by a positive serologic titer to the virus. Immunohistochemistry is highly specific, but it may not be routinely available. Samples for Confirmation of Diagnosis • Virology—lung, mammary gland, synovial membrane, brain (PCR, ISO) • Bulk or individual milk—(PCR, ELISA) • Serology—heart blood serum (AGID, ELISA, PCR) • Histology—formalin-fixed lung, bronchial lymph node, mammary gland, synovial membrane, half of brain section midsagittally (LM, IHC)

DIFFERENTIAL DIAGNOSIS There are several chronic pneumonias requiring differentiation from maedi: • Jaagsiekte • Parasitic pneumonia • Chronic suppurative pneumonia • Caseous lymphadenitis • Postdipping pneumonia • Enzootic pneumonia • Melioidosis • Chronic wasting conditions: • Johne’s disease • Caseous lymphadenitis

TREATMENT No treatment has been successful. Secondary bacterial infections can be treated with commonly used antibiotics, such as tetracyclines, but there will be no improvement in the underlying chronic pneumonia.

CONTROL

In the past, the only control attempted was eradication by complete destruction of all sheep in a flock or area and subsequently restocking. However, it is possible to greatly reduce the prevalence, and even eradicate the disease, by either (a) testing all sheep and removing seropositive sheep from the flock, or (b) by removal of lambs at birth and rearing them in isolation from other sheep. Many jurisdictions have developed accreditation programs for flocks to establish that they have a low risk of infection with MVV. Once flocks are seronegative they are subjected to testing at various intervals, typically 1 to 3 years depending on an assessment of the biosecurity risk and the presence of untested sheep on the same farm holding.

Diseases of the Ovine and Caprine Respiratory Tract

VetBooks.ir

Test and Cull Test and cull involves the detection and culling of seropositive animals and is the preferred method when lateral transmission is the dominant mode of transmission in the flock. All sheep (and goats) on the farm are serologically tested once or twice a year, and seropositive animals and their progeny of less than 1 year of age are removed (culled), preferably for slaughter. If immediate slaughter is not feasible, the seronegative flock must be kept isolated from infected sheep and clothing and equipment that have been in contact with any seropositive animals. Testing is continued semiannually or annually until there are at least two consecutive negative tests. The offspring of older sero­ negative ewes are kept for replacements. Using this approach, an initial seroprevalence to MVV of 66% in a Spanish dairy flock was reduced to 0.2% within 2 years and remained below 2.2% for the next 4 years, and the seronegative flock returned to pretest numbers within 8 years.13 Testing all animals, not just those greater than 1 year old, using a combination of serology and real-time PCR assay to detect proviral DNA, combined with a shorter testing interval of 3 months, may be able to accelerate eradication.14 Using this system, antibody and proviral DNA–negative ewes, proviral DNA–negative lambs, and antibody and proviral DNA–negative yearling ewes were retained as breeders. The PCR test can discriminate lambs that are not infected but serologically positive as a result of maternal antibodies. Segregated Rearing Lambs must be separated from the ewes at birth and receive no colostrum from their dam. They can be given bovine colostrum, or colostrum from a known seronegative flock, then reared on milk replacer completely separate from other sheep. This method may be of particular value when lines of sheep of high genetic merit are desirable to maintain. The disadvantage is that it is labor-intensive and expensive, and there is no cash flow unless the infected sheep are maintained in production pending the establishment of a mature infection-free flock. However, retaining infected sheep creates considerable potential for reinfection of the artificially reared flock, either via accidental contact or fomites. With either method, any future introductions into the flock should be sourced from a known seronegative flock. Flock Biosecurity and Other Control Methods Once infection is introduced it is difficult and expensive to eradicate; thus, establishing and maintaining good biosecurity is a cost-effective way of preventing the introduction of maedi-visna and other important infectious diseases, such as foot

rot. Unfortunately, the specificity and sensitivity of most currently available serologic tests are inadequate to reliably determine the infection status of an individual. Con­ sequently, the results of flock tests from a potential source of replacement sheep should be examined, along with postmortem and other animal health records if these are available. Rams and replacement ewes should be acquired from accredited free flocks in countries where these programs exist and should be transported directly from the source farm rather than through markets or farms of unknown status. Other control procedures that attempt to limit or delay the spread of infection and, consequently, the occurrence of clinical disease within an infected flock have limited success. Lambing in sheds and close confinement paddocks is conducive to spread of disease, and so ceasing or modifying this practice is recommended for infected flocks. In flocks that have a high incidence of clinical disease, culling animals before the age at which they develop clinical signs can reduce the economic impact of the disease. In countries where the disease is endemic, there is often a great deal of movement of animals between farms, especially rams but also replacement ewes. Thus restricting movement of animals between farms and preventing comingling in common grazed areas should help limit the spread of the disease. Vaccination and Genetic Selection There is currently no effective vaccine against the SRLVs, including MVV and OPPV, and in some cases candidate vaccines have enhanced viremia and/or the immunemediated pathology of the disease.2 The difficulty in developing effective vaccines is common among the lentiviruses, with various approaches, including attenuated vaccines, vector vaccines, and proviral DNA vaccines, having little success. The reasons are obscure but probably relate to the underlying dysfunction in T-cell-mediated immune responses. However, marker-assisted genetic selection, to identify those sheep less susceptible to infection, has the potential to supplement existing control measures. For example, in a trial involving 187 lambs, the probability of infection following natural exposure to OPPV was 3.6 times greater in crossbred lambs with susceptible or heterozygous diplotype to ovine transmembrane protein gene 154 (TEM154 diplotype “1 3” or “3 3”) compared with lambs with diplotype “1 1.”15 This is an active research area, and it is expected that additional markers will be identified with additional investigations. FURTHER READING Blacklaws B. Small ruminant lentiviruses: immunopathogenesis of visna-maedi and caprine arthritis and encephalitis virus. Comp Immunol Infect Dis. 2012;35:259-269.

977

Bowles D. Recent advances in understanding the genetic resources of sheep breeds locally-adapted to the UK uplands: opportunities they offer for sustainable productivity. Frontiers Genetics. 2015;6:24. doi:10.3389/fgene.2015.00024. Patel JR, et al. Important mammalian veterinary viral immunodiseases and their control. Vacc. 2012;30:1767-1781. Radostits O, et al. Ovine progressive pneumonia (maedi, maedi-visna). In: Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1362-1366. White SN, Knowles DP. Expanding possibilities for intervention against small ruminant lentiviruses through genetic marker-assisted selective breeding. Viruses. 2013;5:1466-1499.

REFERENCES

1. Le Roux C, et al. Curr HIV Res. 2010;8:94. 2. Blacklaws B. Comp Immunol Microbiol Infect Dis. 2012;35:259. 3. Heaton MP, et al. PLoS Genet. 2012;8:e1002467. 4. Heaton MP, et al. PLoS ONE. 2013;e55490. 5. Bowles D, et al. PLoS ONE. 2014;9:e87823. 6. Gerstner S, et al. JAVMA. 2015;247:932. 7. Fournier D, et al. Can Vet J. 2006;47:460. 8. Ritchie C, Hosie B. Vet Rec. 2014;175:50. 9. Christodouloplous G. Small Rumin Res. 2006;62:47. 10. de Andres X, et al. Vet Immunol Immunopathol. 2013;152:277. 11. Brinkhof J, et al. Small Rumin Res. 2006;70:194. 12. Brinkhof JMA, et al. Vet Microbiol. 2010;142:193. 13. Pérez M, et al. Prev Vet Med. 2013;112:423. 14. Brinkhof JMA, et al. Res Vet Sci. 2010;88:41. 15. Leymaster KA, et al. J Anim Sci. 2013;91:5114.

OVINE PULMONARY ADENOCARCINOMA (JAAGSIEKTE, PULMONARY ADENOMATOSIS) SYNOPSIS Etiology Jaagsiekte sheep retrovirus. Epidemiology Disease of mature sheep with geographic clustering but low prevalence. Spread probably mainly by respiratory route Key signs Dyspnea, profuse watery pulmonary discharge, loud fluid sounds on auscultation, long clinical course with progressive emaciation. Pathology Tumors in lung. Diagnostic confirmation Histologic changes are diagnostic and histopathologic confirmation, including immunohistochemistry, is the only method currently available. Treatment None. Control Culling and strict biosecurity.

Jaagsiekte is Afrikaans for “driving disease” because of the tendency for affected sheep to show clinical signs when driven. The disease manifests clinically as a chronic progressive pneumonia and is a contagious disease of sheep resulting from the development of a bronchioalveolar adenocarcinoma in the lungs.

978

Chapter 12 

■ 

Diseases of the Respiratory System

ETIOLOGY

VetBooks.ir

The disease is associated with an infectious beta-retrovirus, jaagsiekte sheep retrovirus (JSRV) of the family Retroviridae. JSRV has two forms, an exogenous infectious form that alone can produce the disease and an endogenous RSRV-related provirus that is present in all sheep genomes.1 The disease has been transmitted experimentally with partially purified retrovirus from infected lungs, by infection with cloned JSRV, and supportive evidence for retrovirus as the causative agent includes an inverse dose relationship between reverse-transcriptase activity in the infectious inoculum and the incubation period of the experimental disease. The presence of retrovirus has been demonstrated in the lungs of sheep with jaagsiekte in different countries, there is serologic cross-reactivity, and strains from different countries have been sequenced. A herpesvirus has also been isolated in several countries from the lungs of sheep with jaagsiekte, but epidemiologic studies show that it is not the causative agent.

EPIDEMIOLOGY Occurrence The disease has worldwide distribution and is recorded in most countries that have significant sheep populations, with the exception of Australia and New Zealand. Until recently there has been no practical method to detect infected sheep, and estimates of the prevalence of jaagsiekte are largely based on clinical or postmortem observations. The prevalence of the disease appears to vary depending on the breed of sheep and the type of flock management. In most endemically infected flocks, annual losses attributable to jaagsiekte are between 2% and 10%, although the tumor is present in a much higher proportion of the flock, and infection without lesions is also common. Annual mortality can be higher in flocks where the infection has recently been introduced and before the disease becomes endemic. PCR analysis of peripheral blood leukocytes of sheep in infected flocks shows significantly higher rates of nonclinical infection. Prevalence varies between countries, and there can be areas of high prevalence within countries; in Britain, the Borders and the east coast of Scotland, and East Anglia in England, appear to be foci of infection from which other outbreaks arise. The prevalence may be higher than generally recognized; in a biased sample, histologic evidence of jaagsiekte was detected in 25% of cases of pneumonia in sheep submitted to a diagnostic laboratory in Scotland over a 6-year period. In a more recent study, ovine pulmonary adenocarcinoma was confirmed in 0.8% of fallen (culled) adult sheep at a slaughterhouse.2 The disease is also a significant cause of mortality in adult sheep in South Africa and Peru, but it is a minor disease in the United

States and Canada. It occurred in epizootic proportions in Iceland during the same period of time as the maedi-visna epizootic but has been eradicated by a rigorous slaughter policy. Animal and Environmental Risk Factors Mature sheep, 2 to 4 years of age, are most commonly affected, but the disease can occur in younger animals. There are reports of the occurrence of jaagsiekte in goats at very low prevalence rates in India and Greece, and the disease has been experimentally transmitted to goat kids. The lesions produced were small and circumscribed, and goats have low susceptibility to infection. Jaagsiekte has a prolonged clinical course and is uniformly fatal. In some reports there is a greater prevalence of onset of clinical disease in the winter months, but in others there is no seasonal variation in clinical onset. Ewes may show a sudden onset of clinical disease in late pregnancy. The incubation period in natural cases is 1 to 3 years, but it may be as short as 5 to 12 months after experimental transmission. Clinical disease is rare in sheep younger than 2 years and is most common at 3 to 4 years of age. Very rarely, cases occur in lambs 3 to 6 months old, and disease can be reproduced in lambs of this age by challenge of very young lambs. A genetic or familial susceptibility to the disease is suspected. Because of the method of spread, the disease is likely to assume more importance in systems of sheep husbandry where there are significant periods of close contact, as, for example, occurs with intensified lamb-rearing systems. Close housing during the winter is a potent predisposing cause and probably accounted for the occurrence of the disease in epizootic form in Iceland. However, the disease occurs commonly in range sheep in other countries. Sheep that have a combined infection with jaagsiekte and the maedivisna lentivirus have an increased ability to transmit maedi-visna infection, and flocks with the combined infection can suffer high losses from pneumonic disease. Transmission Experimental transmission has been effected by pulmonary or IV injection, or by intratracheal inoculation of infected lung material. The incubation period of the experimental disease in young lambs is much shorter than that in mature sheep. The disease has also transmitted by inhalation of infected droplets when sheep are kept in close contact, and it is assumed that the natural mode of transmission is by droplet infection from respiratory secretions, which are copious in sheep with clinical disease. A longitudinal study of the natural transmission showed that infection established readily and rapidly in young lambs and also horizontally in adult sheep, but that the majority of infected sheep

did not show clinical disease during their commercial life span.

PATHOGENESIS

The virus replicates in the type II pneumocytes in the alveolus. Type II pneumocytes and Clara cells in the terminal bronchioles are transformed, and their growth produces intraalveolar and intrabronchiolar polypoid ingrowths. These cells are surfactantproducing secretory cells, and there is also copious production of fluid. The excessive surfactant-like protein produced in the tumor provides a stimulus for the accumulation of macrophages seen in association with this disease. The adenomatous ingrowths of alveolar epitheliums encroach gradually upon alveolar airspace so that anoxic anoxia occurs. The lesions produced by experimental inoculation are identical with those of the naturally occurring disease.

CLINICAL FINDINGS

Affected sheep are afebrile and show progressive respiratory distress with loss of weight. Clinical signs are not evident until a significant proportion of the lung is compromised by the tumor.1 Occasional coughing and some panting after exercise are the earliest signs, but coughing is not a prominent sign in this disease unless there is concurrent parasitic pneumonia. Subsequently there is emaciation, dyspnea, lacrimation, and a profuse watery discharge from the nose, with death from 6 weeks to 4 months later. A diagnostic test, colloquially known as the wheelbarrow test, in this disease is to hold the sheep up by the hindlegs: in affected animals a quantity of watery mucus (up to about 200 mL) runs from the nostrils. Moist crackles are audible over the affected lung areas and may be heard at a distance, so that a group of affected animals are said to produce a sound like slowly boiling porridge. There is no elevation of body temperature unless there is secondary infection, and the appetite is normal. Advanced cases may have cor pulmonale. Pasteurellosis (Mannheimia haemolytica) is a common complication and often the cause of death.

CLINICAL PATHOLOGY

No immune reaction can be detected in affected animals, and there is no serologic test. Sheep in advanced stages of the disease may show neutrophilia and lymphocytopenia. The pulmonary fluid contains round or spherical clusters of epithelial cells, which have the hyperplasic adenomatous epithelium typical of pulmonary lesions and increased numbers of macrophages. Earlier reports of a consistent elevation in circulating immunoglobulin concentrations have not been substantiated. JSRV can be detected by exogenous JSVR-specific PCR in peripheral blood leukocytes and can be used to demonstrate that JSVR is not present in flocks or regions.3

Diseases of the Ovine and Caprine Respiratory Tract

NECROPSY FINDINGS

VetBooks.ir

Lesions are usually restricted to the thoracic cavity. As in maedi, the lungs are grossly increased in size and in weight (up to 3 times normal). There are extensive areas of neoplastic tissue, particularly of the anterioventral regions of one or both lungs, with smaller lesions in the diaphragmatic lobes. The affected areas are solid and slightly raised above the adjacent normal lung. This, with the excess frothy fluid in the bronchi, is characteristic. The bronchial and mediastinal lymph nodes are enlarged and hyperplastic, and they occasionally contain small metastases. Pneumonic pasteurellosis is a frequent complication, and secondary pulmonary abscesses and pleurisy may develop. Histologically, the alveolus is lined by cuboidal and columnar epithelial cells that form characteristic adenomatous ingrowths of alveolar epithelium into the alveolar spaces. Differences between the pathology of classical (progressive) and atypical (nonprogressive) forms of the disease are seen using immunohistochemistry, with an influx of T-cell subsets and expression of MHC class II in the latter.4 Samples for Confirmation of Diagnosis • Histology—formalin-fixed lung, bronchial lymph node (LM) DIFFERENTIAL DIAGNOSIS Chronic pneumonias requiring differentiation from jaagsiekte: • Maedi • Parasitic pneumonia • Chronic suppurative pneumonia • Caseous lymphadenitis • Postdipping pneumonia • Enzootic pneumonia • Melioidosis

TREATMENT

No treatment is available.

CONTROL

In Iceland, where the disease assumed epizootic proportions, eradication was achieved in the 1950s by complete slaughter of all sheep in the affected areas. In areas where the prevalence is lower, the disease can be satisfactorily controlled, but not eradicated, by slaughter of clinically affected sheep. There is evidence that the disease is spreading in sheep populations in some countries, such as the United Kingdom, and flocks that are free of disease should attempt to obtain replacement sheep from flocks that are free of jaagsiekte. Infected flocks can reduce the prevalence of disease by culling sheep at the onset of clinical signs and also culling the progeny of affected ewes. PCR can detect infection in the preclinical stages, but there has been no trial to establish if eradication

from a flock can be achieved with this technology. Exclusion of the disease from unaffected flocks requires strict biosecurity measures. FURTHER READING Griffiths DJ, Martineau HM, Cousens C. Pathology and pathogenesis of ovine pulmonary adenocarcinoma. J Comp Pathol. 2010;142:260-283. Radostits O, et al. Ovine pulmonary adenocarcinoma (jaagsietke, pulmonary adenomatosis). In: Veterinary Medicine: A Textbook Of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1366-1368.

REFERENCES

1. Scott P, et al. In Pract. 2013;35:382. 2. Cousens C, et al. Vet Rec. 2015;176:413. 3. Maeda N, et al. J Vet Med Sci. 2011;73:1493. 4. Summers C, et al. Vet Immunol Immunopathol. 2012;146:1.

979

Inflammation of these surfaces tissues is evident and increases as the larvae become mature. Changes to the epithelial structure are noted, including the erosion of the surface ciliary covering and a breakdown in epithelial cell integrity. Abrasive action of the body armature and the activity of proteolytic enzymes excreted/secreted by the larvae are responsible for the pathology. Secondary effects Include the induction of lung lesions and the activation of latent “orf” infections. Diagnostic confirmation. Behavioral changes during fly season and nasal discharge Differential diagnosis Unthriftiness usually caused by helminth infection. Treatment Macrocyclic lactone endectocides, clorsulon. Control Treatment given when fly activity has ceased.

NASAL BOTS INFESTATION Infestation of sheep and goats with larvae of the nasal bot fly (Oestrus ovis) has a serious effect on the productivity and welfare of both sheep and goats. Adult activity induces stress responses and significant behavioral change. Larval infestation induces moderate to severe pathology that reduces productivity. Similar flies are known to affect horses, donkeys, and mules (Rhinoestrus spp.) in the Mediterranean region and to affect camels (Cephenemyia titillator) in Africa and Australia. Wild ungulates are affected by nasal bots (e.g., Cephenemyia spp.). Very little is known about the pathology and impact of these later groups of flies, but similarities in life history suggest their effects will be similar to those discussed here. SYNOPSIS Etiology Oestrus ovis inhabits the nasal passages and sinuses of sheep and goats. Similar species affect horses and camels. Epidemiology Larvae are sprayed onto the nares of hosts by passing females. Flies are active during spring and summer, inducing behavioral changes in hosts under attack. In temperate climates there is only a single generation per year, but in warmer climates two generations occur. First instars in the nasal passages undergo hypobiosis during winter or hot summer when survival of pupae or adults is low, resuming development when conditions are more favorable. Clinical signs Shortly after arrival of the larvae an increase in nasal discharge and sneezing are evident. As the infestations develop the amount of discharge increases and the nostrils may become caked with dust and debris, forcing the infested animals to breathe through their mouth. Clinical pathology Changes are noted to the mucosa of the ethmoid and sinus regions.

ETIOLOGY The sheep nose bot affects sheep and goats in most regions, but it is particularly significant in the Mediterranean basin, central America, southern Africa, and eastern Europe. The larvae inhabit the nasal passages and sinuses, eventually being expelled through the nares. Goats are less dramatically affected than sheep. The slightly dorsoventrally flattened, segmented larvae are light cream in color, but as they reach maturity dark bands appear on each segment. Species affecting horses and camels have distributions that are similar.

LIFE CYCLE AND EPIDEMIOLOGY

The adult fly is stout, mottled gray in color, and about 1cm long. Its mouthparts are rudimentary, and it does not feed. In North America, flies emerge in the late spring and mate, and the females begin larviposition activities approximately 2 to 3 weeks later. Adult flies attempting to deposit larvae on the nares annoy the sheep and cause them to bunch or seek shelter. Stamping of the feet and shaking of the head are common. Sheep may bunch together and press their heads into the fleece of others. Fly activity occurs primarily during the warmer parts of the day but still may result in the loss of a good deal of grazing time. Behavioral changes in goats are less dramatic, presumably because of their browsing habit. Larval development takes place in in the dorsal turbinates and frontal sinuses. The period of development can vary 3 weeks to several months, after which they migrate to the nostrils. Larvae feed on the mucosal secretions and cells eroded from the mucosal epithelium. The larvae are thick, yellowishwhite in color, and when mature there is a dark dorsal band on each segment. The ventral surface has rows of small spines on each segment. Mature larvae exit the host, usually during a bout of sneezing, and

980

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

actively burrow beneath the upper layers of soil and ground litter. Pupation occurs at these locations, and development of the adults requires 4 to 5 weeks but may take longer at low temperatures. In temperate areas there may be one or two generations per year, but several generations may be completed in hot areas. O. ovis are adapted to the various climates prevailing wherever sheep and goats are kept. When winters are cold, the larvae can overwinter by remaining dormant in the first instar (hypobiosis), but in warmer climates development may continue throughout the winter. In those regions where summer temperatures are extreme, the larvae will also undergo hypobiosis. O. ovis are an important zoonosis because the females may larviposit in the eye, in the nose, or on the lips of humans. In some countries ophthalmomyiasis or infection of the upper respiratory tract is a common occurrence.

PATHOGENESIS

The stress of the larviposition attacks can be significant with reduced grazing time and overheating resulting from bunching. Herdsmen find the animals are more nervous and difficult during the fly activity periods. Larvae induce a gradually increasing rhinitis and sinusitis as the infestation persists. Marked changes in the structure of the epithelial tissues are noted. with a marked cellular degeneration and a loss of the ciliary layer. The changes are a result of both mechanical activity of the larval spines and mouthhooks and the effect of proteolytic enzymes excreted or secreted.1,2 Varying degrees of mucous discharge are observed in the later stages of the infestation. This can lead to the nostrils being occluded by adherent straw and dust.

CLINICAL FINDINGS

Early in the infestation there is a distinct rhinitis accompanied by a muco- to mucopurulent discharge.1 Later as larvae mature a sinusitis is evident. Presence of mature larvae in nasal cavities may induce excessive sneezing, which assists larval exit. Activity of the larvae in the nasal cavities, and the changes they induce lead to an increases incidence of secondary pathology. The number and severity of lung abcess are more significant in nose bot–infested sheep. The presence of bots also is correlated with increased carcinomas and may lead to reactivation of latent “orf ” symptoms.

DIAGNOSIS

The behavioral changes during fly activity, including bunching and burying of noses in neighbors’ fleeces, is a reliable indicator of fly attack. Nasal discharge and excessive sneezing are highly suggestive but not definitive. Infested sheep and goats develop some level of immunity from exposure to larval antigens but is unlikely to be used on the farm.2

An ELISA for detection of antibodies to larvae secretions has been developed but is not currently used.2

Neostrongylus linearis have been recorded in some countries.

TREATMENT

D. filaria has a direct life cycle like that of D. viviparus in cattle. The life cycles of the other (protostrongylid) species are similar except that they have different predilection sites in the lung and have indirect life cycles with molluscan intermediate hosts. Transmission occurs when infected slugs or snails are accidentally ingested during grazing.

Closantel 5 mg/kg and ivermectin 0.2 mg/ kg, in addition to other macrocyclic lactones, are effective, and the use of these compounds for fluke or worm control also controls nasal bots.

CONTROL

Treatment should preferably be applied after the cessation of fly activity, although it may be necessary to apply treatments during prolonged fly activity to give relief.1

RECOMMENDATION

Treatment should be applied once or twice a year. This is not absolutely necessary but will increase both endurance and the animal’s well-being. Population control of the flies is probably not likely. REFERENCES

1. Angulo-Valadez CE, et al. Med Vet Entomol. 2010;25:117-125. 2. Angulo-Valadez CE, et al. Vet Parasitol. 2010;174:19-25. 3. Panadero-Fontan R, Otranto D. Vet Parasitol. 2015;208:84-93.

LUNGWORM INFESTATION IN SHEEP AND GOATS SYNOPSIS Etiology The nematode parasites Dictyocaulus filaria, Muellerius capillaris, and Protostrongylus rufescens. Epidemiology Infective D. filaria larvae are found on grass, but M. capillaris and P. rufescens are transmitted when molluscan intermediate hosts are accidentally ingested by grazing animals. Signs D. filaria and P. rufescens can cause bronchitis and loss of condition. M. capillaris is asymptomatic in sheep but may be pathogenic in goats. Clinical pathology Characteristic larvae in feces. Lesions D. filaria and P. rufescens: scattered patches of consolidation; M. capillaris: small fibrous nodules up to 5 mm in diameter. Diagnostic confirmation Characteristic larvae in feces. Treatment Avermectins/milbemycins, benzimidazoles or levamisole. Control No specific measures available.

ETIOLOGY

Infestations with the nematode Muellerius capillaris are ubiquitous. Dictyocaulus filaria and Protostrongylus rufescens are encountered sporadically. Cystocaulus ocreatus and

LIFE CYCLE

EPIDEMIOLOGY

D. filaria infestations in sheep appear to follow the same pattern as those of D. viviparus in calves, but the number of lungworms is usually low. The third-stage larvae are long-living in damp, cool surroundings. The lambs of one season are the main source of infection for the next season’s lambs, but larvae passed by ewes and yearlings also contribute to pasture contamination. The prevalence of infection is low in spring and summer but rises rapidly in the autumn and winter, when most clinical cases are seen. Warm, wet summers give rise to heavier burdens in the following autumn and winter. Immunity after natural exposure is strong and durable in sheep but less so in goats. M. capillaris infestations in sheep have been recorded from most parts of the world,1,2,3 and in many temperate areas almost all sheep are infected.4,5 Massive invasion with larvae is uncommon because the intermediate hosts are not usually ingested in large numbers nor are they grossly infested with larvae. Massive infestations with this worm do not develop acutely, and heavy infestations, when they occur, appear to develop over a long period of time. Infected sheep carry patent infection from 1 year to the next.

PATHOGENESIS

The relative pathogenicity of each lungworm is dependent on its predilection site. D. filaria lives in the trachea and bronchi so aspirated eggs, larvae, and debris can affect a large volume of lung tissue. It is therefore the most pathogenic species and provokes changes resembling those described for D. viviparus. The volume of damaged lung is however usually insufficient to cause severe dyspnea. Adult P. rufescens are found in smaller bronchioles, and so associated lesions are much smaller. M. capillaris is found in the lung parenchyma, where it becomes encysted in fibrous nodules. Lesions are thereby confined to its immediate surroundings. Consequently, this worm is generally considered to be relatively innocuous. Heavy mixed protostrongylid infections can impair pulmonary gaseous exchange.

CLINICAL FINDINGS

Lambs 4 to 6 months of age are most severely affected with lungworms, but sheep of all

Diseases of the Equine Respiratory Tract

VetBooks.ir

ages are susceptible. Clinically D. filaria is associated with bronchial irritation that results in coughing, moderate dyspnea, and loss of condition. There may be added fever and evidence of toxemia if secondary bacterial infection occurs. It is highly pathogenic in young goats. P. rufescens infestations in sheep and goats cause clinical signs similar to those of D. filaria.

CLINICAL PATHOLOGY

Laboratory diagnosis depends on the detection of first-stage larvae in the feces by the Baermann technique. D. filaria larvae have refractile granules in their intestinal cells and a conical tail. P. rufescens has a wavy tail as does M. capillaris, which, in addition, has a spine just anterior to the tail.

NECROPSY FINDINGS

D. filaria lesions are similar to those of the subacute form of parasitic bronchitis in calves with exudate in the bronchioles, scattered patches of consolidation, and thickening of the alveolar septa,6-8 but widespread lesions are not common. M. capillaris is found in small fibrous nodules up to 5 mm in diameter. Most of these are in the parenchyma of the lung immediately under the pleura. Many of them are calcified and often contain only one live or dead worm. Infestation of goats leads to a diffuse infection quite different from the nodular reaction in sheep and to the production of an interstitial pneumonia. Whether this is attributable solely to M. capillaris infection or whether a chlamydial or viral agent is involved has not been determined. However, cases of nodular reaction in goats attributable to M. capillaris larvae have been reported.6

DIAGNOSTIC CONFIRMATION

The presence of larvae in the feces confirms lungworm infection, but their number is often no indication of the degree of infestation. DIFFERENTIAL DIAGNOSIS Lungworm infestation in sheep needs to be differentiated from maedi and jaagsiekte.

TREATMENT TREATMENT Ivermectin (0.2 mg/kg, SC) (R-1) Moxidectin (0.2 mg/kg, SC or PO) (R-1) Fenbendazole (5 mg/kg PO, every day for 7 days) (R-2) Albendazole (7.5 mg/kg BW, PO) (R-2)

Ivermectin, moxidectin, the benzimidazoles, and levamisole are effective against D. filaria at normal dose rates. Ivermectin, in addition, has a label claim for P. rufescens. It is doubtful whether treatment of sheep for M. capillaris

is ever justified. In goats, one or two doses of ivermectin (0.2 mg/kg, SC o rPO) or elevated doses of benzimidazoles destroys the adult worms but not the immature stage, but regular daily oral doses of fenbendazole (up to 5 mg/kg/d) in the feed for 1 to 2 weeks or albendazole (1 mg/kg in the feed for 2 weeks) are highly effective against all stages. The label dose of albendazole (7.5 mg/kg BW, once in sheep and 10 mg/kg BW once in goats) is effective in treating adult lungworms, but is not effective against the immature stage.

CONTROL

An attenuated vaccine for D. filaria is available in a few countries where this worm is a particular problem. With most forms of sheep husbandry, there are few precautionary measures that can be taken, particularly against lungworms with molluscan intermediate hosts. REFERENCES

1. Borji H, et al. Asian Pac J Trop Med. 2012;5:853. 2. Nematollahi A, et al. J Vet Res. 2009;64:339. 3. Regassa A, et al. Vet Parasitol. 2010;169:144. 4. Domke AV, et al. Vet Parasitol. 2013;194:40. 5. Kouam MK, et al. Vet J. 2014;202:146. 6. Panayotova-Pencheva MS, et al. Vet Med Int. 2010;2010:741062. 7. Yildiz K, et al. Helminthologia. 2006;43:208. 8. Iacob O, et al. Sci Parasitol. 2007;1:72.

Diseases of the Equine Respiratory Tract ABNORMALITIES OF THE UPPER RESPIRATORY TRACT OF HORSES Impairment of ventilation by abnormalities of the upper respiratory tract is an important cause of poor performance in athletic horses. Abnormalities that impair athletic capacity are those that reduce the effective diameter of the upper airway, thereby increasing the work needed to maintain the same level of tidal volume and minute ventilation or, as is the case clinically, reducing the minute ventilation achieved by the horse during maximal exercise. In other words, a reduction in effective diameter of the upper airway increases work of breathing at all exercise intensities, and at maximal intensity, when the effort expended on breathing cannot be increased, decreases maximal minute ventilation. The result is diminished oxygenation of arterial blood and delivery of oxygen to muscle and other tissues, exacerbated hypercapnia, and reduced athletic capacity during high-speed exercise.1-3 The work of breathing is, simplistically, determined by the volume of air moved and the pressure required to do so. The relationship between pressure and resistance in the airway is described mathematically by a rearrangement of Poiseuille’s law:

981

Resistance to flow = pressure drop flow = 8 × viscosity of the air × length of the airway (π × [radius]4 ) Given that the viscosity of air is constant and the length of the airway does not change for an individual horse, the radius of the airway has a huge effect on resistance to flow. Notice that a change in pressure is inversely proportional to the fourth power of the radius (r4), with the consequence that relatively small changes in radius have large effects on the pressure needed to generate a given flow of air. For this reason, abnormalities of the upper airway that cause only small reductions in airway diameter can have clinically important effects on ventilation during high intensity exercise. Another consequence of changes in airway diameter and structure is the generation of abnormal airflow patterns that result in production of abnormal respiratory sounds. Such sounds can vary from gurgling through to roaring and can be of diagnostic importance.4 Advent of first rigid and then flexible endoscopes allowed greater refinement of diagnosis of disorders of the upper respiratory tract of horses when examined at rest. A further advance was the ability to examine the upper airway during intense exercise. This was first achieved in horses running on a treadmill and has now progressed to examination of horses running over ground. Although there are advantages to each mode of examination (at rest, treadmill, over ground), greatest diagnostic utility is achieved by examination of horses exercising over ground and performing their customary activity wearing their usual tack and with their rider.5-8 Laryngeal hemiplegia caused by recurrent laryngeal neuropathy is a wellrecognized abnormality of the upper airway associated with impaired performance by racehorses. In many of its forms, it is readily identified in horses at rest. However, more subtle abnormalities or those that develop as the horse fatigues are best detected, or can only be detected, on examination during strenuous exercise. It is now clear that most abnormalities of the upper airway of horses, with the exception of laryngeal hemiplegia, can only reliably be detected by examination of exercising horses.9 Abnormalities developing during strenuous exercise by horses are best referred to as “dynamic” abnormalities. This term should not be used to denote the mode of examination (ie, “dynamic endoscopy”), which should be specified as “over ground” or “treadmill.” Terms describing abnormalities detected during endoscopic examination of the upper airway of horses have recently been standardized (Table 12-10).10 The use of endoscopy during exercise has revealed that dynamic abnormalities of the upper airway are often complex and involve

982

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Table 12-10  Preferred terms for describing findings on endoscopic examination of the upper airway of horses (modified from10)

Preferred term

Preferred abbreviation

Also known as

Recurrent laryngeal neuropathy

RLN

Laryngeal paralysis, laryngeal hemiplegia

Dynamic laryngeal collapse

DLC

Bilateral arytenoid cartilage collapse

Intermittent dorsal displacement of the soft palate

iDDSP

Persistent dorsal displacement of the soft palate

pDDSP

Palatal instability

PI

Vocal fold collapse

VFC

Vocal cord collapse

Medial deviation of the aryepiglottic fold

MDAF

Aryepiglottic fold collapse, Axial deviation of the aryepiglottic fold

Nasopharyngeal collapse

NPC

Nasopharyngeal obstruction, pharyngeal wall collapse

Ventromedial luxation of the apex of the corniculate process of the arytenoid

VLAC

Collapse of the apex of the corniculate process of the arytenoid

Permanent DDSP

Cricotracheal ligament collapse Collapse of the margins of the epiglottis Epiglottic retroversion Rostral deviation of the palatopharyngeal arch

RDPA

High speed treadmill endoscopy

HSTE

Overground endoscopy

OGE

multiple structures.9,11 Up to 50% of horses examined during high-speed exercise have multiple abnormalities of the upper airway. Furthermore, examination of exercising horses has revealed abnormalities not apparent during examination of resting horses, including the full spectrum of manifestations of recurrent laryngeal neuropathy, palatal instability including intermittent dorsal displacement of the soft palate, vocal fold collapse, aryepiglottic fold collapse, axial deviation of the aryepiglottic folds, dynamic nasopharyngeal collapse, collapse of the corniculate process of the arytenoid cartilage, bilateral arytenoid and vocal fold collapse, and epiglottic retroversion.9 FURTHER READING Franklin SH, Allen KJ. Assessment of dynamic upper respiratory tract function in the equine athlete. Equine Vet Educ. 2015;doi:10.1111/eve.12432.

REFERENCES

1. Davidson EJ, et al. J Equine Vet Sci. 2011;31:475. 2. Courouce-Malblanc A, et al. Equine Vet J. 2010;42:246. 3. Allen K, et al. Equine Vet J. 2013;45:350. 4. Burn JF, et al. Equine Vet J. 2006;38:319. 5. Allen KJ, et al. Equine Vet J. 2010;42:186. 6. Allen KJ, et al. Equine Vet J. 2010;42:587. 7. Van Erck E. Equine Vet J. 2011;43:18. 8. Kelly PG, et al. Equine Vet J. 2013;45:700. 9. Barakzai SZ, et al. Equine Vet J. 2012;44:501. 10. Barnett TP, et al. Equine Vet J. 2015;47:505. 11. Van Erck-Westergren E, et al. Equine Vet J. 2013;45:376.

Dynamic respiratory endoscopy, telemetric endoscopy

PALATAL DYSFUNCTION (INSTABILITY, DORSAL DISPLACEMENT OF THE SOFT PALATE) The soft palate of equids is unique in that it provides an airtight seal between the oropharynx and nasopharynx during respiration, rendering equids obligate nasal breathers. During swallowing the soft palate is transiently displaced dorsally to permit passage of the feed bolus as part of the normal act of deglutition. Abnormalities of the soft palate that result in alteration of its anatomic position (displacement) or inability to maintain normal tone during exercise are associated with impaired respiration and exercise intolerance.1-3 Displacement of the soft palate other than during deglutition is abnormal and can be intermittent, which is usually associated with exercise, or persistent, which is usually associated with disruption of the nerve supply to the pharynx. Palatal Instability and Intermittent Dorsal Displacement of the Soft Palate During Exercise Dysfunction of the palate during exercise results in a range of structural abnor­ malities that reduce the functional area of the rima glottidis (the opening of the larynx) and thus impair ventilation during high-speed exercise. Dysfunction during exercise ranges from palatal instability to

Fig. 12-18  Endoscopic view of dorsal displacement of the soft palate in a resting Thoroughbred racehorse.

intermittent dorsal displacement of the soft palate.4 Palatal instability and dorsal displacement of the soft palate cause an expiratory obstruction to airflow through the larynx and pharynx.4 Palatal instability is evident as dorsoventral billowing of the soft palate during high speed exercise and flattening of the epiglottis against the palate. Palatal instability is significantly associated with dorsal displacement of the soft palate during exercise.1 Dorsal displacement of the soft palate is an extreme of palatal instability and results when the caudal border of the soft palate displaces dorsal to the epiglottis (Fig. 12-18). Palatal instability is also associated with axial deviation of the aryepiglottic folds and abnormalities in the conformation of the epiglottis.1,5 Estimates of the prevalence of the disease in the wider population are unreliable because of the transient nature of the instability and displacement and the fact that it only occurs during exercise. Additionally, examination of large numbers of horses to determine prevalence in populations of horses has not been performed, with most reports being prevalence rates in horses selected for high-speed endoscopic examination. It is estimated to occur in 0.5% to 1.3% of Thoroughbred racehorses, and of 52 Thoroughbred racehorses examined using over ground endoscopy, 25% had dorsal displacement of the soft palate, 40% had axial deviation of the aryepiglottic fold, 35% had vocal fold collapse, and 33% had abnormal arytenoid function.6 Forty-eight percent of the horses had multiple abnormalities. Nineteen of 57 Thoroughbred yearlings had intermittent dorsal displacement during a single examination using over ground endoscopy.7 Dorsal displacement was detected in 10 of 46 Standardbred racehorses examined using overground endoscopy during racing—these horses were presumably considered healthy before examination.8 Three

Diseases of the Equine Respiratory Tract

VetBooks.ir

percent of performance horses (nonracing) had dorsal displacement of the soft palate as the sole abnormality during exercise.9 Palatal instability and intermittent dorsal displacement of palate are a common part of complex dynamic abnormalities of the upper respiratory tract in harness horses, with 7 0% of examined horses having a complex disorder.5 Similarly, 19% of performance horses (nonracing horses) had complex upper respiratory tract abnormalities during exercise.9 The cause of intermittent displacement of the soft palate during exercise is unknown, although a number of mechanisms, including palatal myositis, ulcers of the caudal border of the soft palate, caudal retraction of the larynx, and lower respiratory disease, are suggested. Retropharyngeal lymphadenopathy can cause neurogenic paresis of the pharyngeal and palatal muscles, with dorsal displacement of the soft palate the most obvious sign of pharyngeal collapse during exercise. The immediate cause of the displacement is the high turbulent flow and negative intrapharyngeal pressure generated during exercise.10 Displacement of the soft palate during strenuous exercise places the soft palate dorsal to the epiglottis, a position in which it impedes flow of air during expiration. Peak expiratory airflow, minute ventilation, tidal volume, and rate of oxygen consumption are all decreased in horses with dorsal displacement of the soft palate, whereas inspiratory flow and breathing rate are not affected.4 Clinical Signs The clinical signs include exercise intolerance and intermittent production of a gurgling noise during strenuous exercise. Endoscopic examination of resting horses usually demonstrates a normal pharynx and larynx. Brief nasal occlusion (30–60 s) that induces displacement of the soft palate (Fig. 12-18), in combination with a history of respiratory noise during exercise, increases the likelihood of the disorder. Endoscopic examination of affected horses during exercise is the gold standard for diagnosis and reveals signs of palatal instability or dorsal displacement of the soft palate and related abnormalities. Detection of palatal instability and associated abnormalities is described as follows:1 Axial deviation of the aryepiglottic folds: Graded as none, mild, moderate or severe. 1. Mild ADAF, defined as axial collapse of the aryepiglottic folds with the folds remaining abaxial to the vocal cords. 2. Moderate ADAF, defined as axial deviation of the aryepiglottic folds less than halfway between the vocal cord and the midline.

3. Severe ADAF, defined as collapse of the aryepiglottic folds more than halfway between the vocal cord and the midline. Epiglottic conformation: Epiglottic conformation is categorized into three groups. 1. Convex epiglottic appearance when the epiglottis maintained a convex shape during exercise; typically only the tip of the epiglottis is in contact with the soft palate. 2. Flattened epiglottis where the epiglottis loses its convex shape and appears to lie flat or slightly concave on the surface of the soft palate, but the tip of the epiglottis remains ventral to the base. 3. A tipped up appearance when the epiglottis has a flattened or concave appearance and during inspiration the tip of the epiglottis is at the same level as or higher than the base of the epiglottis. Obstruction of the rima glottidis by the soft palate (soft palate stability): The stability of the soft palate is graded according to whether the rima glottidis is obscured by the billowing soft palate. 1. The soft palate is considered stable when there is no movement or lifting of the soft palate was observed (Fig. 12-19). 2. Palatal instability with no rima glottidis obstruction when the soft palate lifts up to the level of the base of epiglottis but the rima glottidis is not obscured (Fig. 12-20). 3. Palatal instability with rima glottidis obstruction when the soft palate lifts so that the rima glottidis becomes obscured (Fig. 12-21).

983

epiglottis or billowing dorsally either side of the epiglottis. The presence or absence of a sling appearance to the ventrolateral pharyngeal walls at the level of the guttural pouch ostia should be noted. The caudal soft palate should be assessed as to whether a concave appearance was present and if so should be graded as absent, small, or large during each of inspiration and expiration. Radiographic examination of the pharynx reveals a shortened epiglottis (10,000 cells/µL, 10 × 10 cells/L) comprised principally of degenerative neutrophils, and an increased protein concentration (>2.5 g/dL, 25 g/L), and it may contain intracellular and extracellular bacteria. A Gram stain of the fluid should be examined. The pleural fluid should be cultured for aerobic and anaerobic bacteria. A putrid odor suggests infection by anaerobic bacteria. Sterile pleural fluid has a pH, Po2 and Pco2, and lactate, glucose, and bicarbonate concentration similar to that of venous blood. Infected pleural fluid is acidic and

993

hypercarbic and has an increased concentration of lactate and decreased concentrations of bicarbonate and glucose compared with venous blood. Tracheal aspirates have a leukocytosis comprised of degenerate neutrophils with intra- and extracellular bacteria. Cultures of tracheal aspirates more frequently yield growth than do cultures of pleural fluid (90% vs. 66%).

DIAGNOSTIC CONFIRMATION

The presence of excessive pleural fluid containing bacteria and degenerate neutrophils in combination with clinical signs of respiratory disease provides confirmation of the disease. DIFFERENTIAL DIAGNOSIS Diseases that may cause respiratory distress and pleural effusion in horses include the following: • Intrathoracic neoplasia, including mesothelioma, lymphoma, and extension of gastric squamous-cell carcinoma • Penetrating chest wounds • Esophageal perforation • Diaphragmatic hernia • Congestive heart failure • Hemangiosarcoma (causing hemothorax) • African horse sickness • Pulmonary hydatidosis • Pulmonary infarction and pneumonia

NECROPSY FINDINGS The pneumonia involves all areas of the lungs but is most severe in the cranial and ventral regions. The pleura are thickened and have adherent fibrin tags, and there is excessive pleural fluid. The pleural fluid contains strands of fibrin and is usually cloudy and serosanguineous to yellow. Histologically, there is a purulent, fibrinonecrotic pneumonia and pleuritis.

TREATMENT

Given early recognition of the disease and prompt institution of appropriate therapy, the prognosis for horses with pleuropneumonia is favorable. However, the long course of the disease and the associated expense often limit therapeutic options and make the outcome a decision based on economic rather than medical grounds. The principles of treatment are prompt broad-spectrum antimicrobial therapy; removal of infected pleural fluid and cellular debris, including necrotic lung; relief of pain; correction of fluid and electrolyte abnormalities; relief of respiratory distress; treatment of complications; and prevention of laminitis. Antimicrobial Treatment The prompt institution of systemic, broad-spectrum antimicrobial therapy is the single most important component of

994

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

treatment of horses with pleuropneumonia. Antimicrobial therapy is almost always started before the results of bacterial culture of pleural fluid or tracheal aspirate are received and the antimicrobial sensitivity of isolated bacteria are determined. Use of antibiotics or combinations of antibiotics with a broad spectrum of antimicrobial activity is important because of the polymicrobial nature of most infections and because the wide range of gram-positive and gramnegative bacteria that may be associated with the disease makes prediction of the susceptibility of the causative organisms difficult. Furthermore, superinfection with bacteria, especially Enterobacteriaceae and obligate anaerobes, commonly occurs in horses with disease initially associated with

a single bacterial species. Administration of drugs that are effective in the treatment of penicillin-resistant obligate anaerobes is also important. Recommended doses for antimicrobials used in the treatment of pleuropneumonia are provided in Table 12-11. Antimicrobial therapy should be broad spectrum to include coverage of the likely bacteria involved in the disease. It should therefore provide coverage against Streptococcus spp., Actinobacillus/ Pasteurella spp., Enterobacteriaceae, and anaerobes, including Bacteroides spp. A combination of penicillin G, an aminoglycoside, and metronidazole provides broadspectrum coverage and is a frequently used empirical therapy until the results of bacterial culture are known. Results of bacterial

culture and subsequent antimicrobial susceptibility testing may aid selection of further antimicrobials. However, superinfection with gram-negative and anaerobic bacteria is common, and there is a sound rationale for continued use of a combination of antimicrobials providing broad-spectrum coverage throughout treatment of the disease. Antimicrobial therapy will be prolonged in most cases, usually being required for at least 1 month and often several months. As the disease resolves it may be possible to change from parenteral antibiotics to orally administered antibiotics such as a combination of trimethoprim–sulfonamide, although the clinical response to this combination is sometimes disappointing, or doxycycline or enrofloxacin.

Table 12-11  Antimicrobial agents and recommended doses for treatment of pleuropneumonia in horses

Drug

Dose, route, and interval

Comments

Procaine penicillin G

22,000–44,000 IU/kg IM q12h

Effective against Streptococcus sp. and most anaerobes, with the exception of Bacteroides fragilis. Achieves low plasma concentrations but has prolonged duration of action. Cheap. Synergistic with aminoglycosides. Should not be used as sole treatment.

Sodium or potassium penicillin G

22,000–44,000 IU/kg IV q6h

Effective against gram-positive organisms (except penicillinase-producing bacteria such as Staphylococcus spp.) and most anaerobes. Achieves high plasma concentrations. Synergistic with aminoglycosides. Expensive.

Ampicillin sodium

11–22 mg/kg IV or IM q6h

Wider spectrum than penicillin G. Achieves high plasma concentrations.

Ampicillin trihydrate

20 mg/kg IM q12–24h

Synergistic with aminoglycosides Low blood concentrations. Muscle soreness. Not recommended.

Ceftiofur sodium

2.2–4.4 mg/kg IM or IV q12h

Wide spectrum of action against gram-positive and gram-negative organisms and most anaerobes. Can be used as sole treatment, though not recommended.

Ceftiofur crystalline

7 mg/kg IM q4 days

Prolonged concentration in blood and bronchoalveolar lavage fluid.

Cefotaxime

40 mg/kg IV q6h

Wide spectrum of action against gram-positive and gram-negative organisms and most anaerobes. Can be used as sole treatment, though not recommended.

Cefepime

2.2 mg/kg IV or IM q8h

Wide spectrum of action against gram-positive and gram-negative organisms and most anaerobes. Can be used as sole treatment, although not recommended.

Chloramphenicol

50 mg/kg, PO q6h

Good spectrum of action, including anaerobic bacteria. Poor oral bioavailability and disappointing clinical efficacy. Use prohibited in some countries. Potential human health hazard. Risk of diarrhea

Gentamicin sulfate

7 mg/kg, IV or IM q24h

Active against Staphylococcus spp. and many gram-negative organisms. Inactive against anaerobes. Poor activity against Streptococcus spp. Synergistic with penicillin

Enrofloxacin

7 mg/kg IV or PO q24h

Active against some gram-positive and gram-negative bacteria. Not good or reliable activity against streptococci. Contraindicated in young animals because of risk of cartilage damage.

Amikacin sulfate

10 mg/kg IV or IM q24h

Wider spectrum of gram-negative activity than gentamicin. Expensive

Trimethoprim– sulfonamides

15–30 mg/kg PO q12h

Theoretical wide spectrum of action. Disappointing clinical efficacy.

Rifampin

5–10 mg/kg PO q12h

Penetrates abscesses well. Active against gram-positive and some gram-negative bacteria. Must be used in conjunction with another antibiotic (not an aminoglycoside).

Doxycycline

10 mg/kg PO q12h

Broad spectrum of activity, but resistance unpredictable. Only moderate blood concentrations. Suitable for prolonged therapy but not treatment of the acute disease.

Ticarcillin–clavulanic acid

50 mg/kg IV q6h

Broader spectrum of gram-negative activity than penicillin G. Expensive.

Metronidazole

15–25 mg/kg PO q6–8h

Active against anaerobes only. Used in conjunction with other antimicrobials (especially penicillin and aminoglycosides). Neurotoxicity rare.

Clinical results sometimes disappointing

IM, intramuscularly; IV, intravenously; PO, orally; q, dose administered every “h” hours.

Diseases of the Equine Respiratory Tract

VetBooks.ir

The decision to discontinue antimicrobial therapy should be based on lack of fever, nasal discharge, and respiratory distress or cough; lack of evidence of intrathoracic abscesses on ultrasonographic and radiographic examination of the thorax; and resolution of neutrophilia and hyperfibrinogenemia. There should be no appreciable pleural fluid on ultrasonographic examination. Thoracic Drainage Chronic, effective drainage of the pleural cavity and intrathoracic abscesses is critical for successful treatment of horses with pleuropneumonia. Horses with sterile pleural fluid may require only a single drainage of pleural fluid. More severely affected horses may require intermittent drainage on each of several days, and most cases will require insertion of a tube into the pleural space to provide continuous drainage for several days to several weeks. Horses with chronic disease may benefit from a thoracotomy that provides continuous drainage and the ability to lavage the chest. Ultrasonographic examination of the chest is very useful in identifying the presence of pleural fluid, the optimal sites for drainage, and the efficacy of drainage. Intermittent thoracic drainage can be achieved by inserting a bovine teat cannula or similar blunt cannula into the pleural space. This should be done aseptically and under local anesthesia. If ultrasonographic examination is not available, the cannula should be placed in the sixth to eighth intercostal space on the right side or the seventh to ninth on the left side just above the level of the olecranon. Pleural fluid that does not contain large fibrin clots (which clog the cannula) can be drained and the cannula removed. However, the process is slow if large quantities of fluid must be removed. Intermittent drainage is indicated when the quantities of pleural fluid are small (< 5 L), relatively cell-free, or localized. This situation is most likely to occur in horses with acute disease. Insertion of large plastic chest tubes (20–30 French, 6- to 10-mm outside diameter) facilitates rapid fluid removal, allows drainage of viscid fluid, and provides continuous drainage. The chest tube should be inserted in an aseptic fashion under local anesthesia at sites indicated by ultrasonographic examination or as described previously. A one-way valve should be attached to the external end of the tube to prevent aspiration of air and development of a pneumothorax. A balloon or condom with the end removed is an effective one-way valve. The chest tube is secured to the chest wall with a purse-string suture. The tube may be retained for several days to a week, but it should be monitored frequently (every few hours) and cleared of fibrin clots as needed. Complications of drainage of pleural fluid include collapse of the animal if the

fluid is removed too rapidly, pneumothorax, sudden death as a result of cardiac puncture or laceration of a coronary vessel, and perforation of abdominal viscera. Collapse can be prevented by administering fluids intravenously during pleural fluid drainage and by removing the fluid gradually (over a period of 30 minutes). Some horses develop cellulitis around the chest tube, which requires that the tube be removed. Thoracotomy may be required in recurrent or chronic cases to provide drainage of intrathoracic abscesses or chronic pleural effusion that is refractory to treatment with antimicrobials. Thoracotomy is an effective intervention in many horses, with 14 of 16 horses treated by thoracotomy surviving and 6 returning to athletic activity.8 Thoracotomy should not be considered an emergency or heroic procedure in such cases. Pleural Lavage Infusion and subsequent removal of 5 to 10 L of warm saline or balance polyionic electrolyte solution into the affected pleural space may be beneficial in the treatment of cases with viscid fluid or fluid containing large amounts of fibrin and cell debris. The fluid can be infused through the chest tube that is used to drain the pleural space. Care should be taken not to introduce bacteria with the infusion. Fibrinolytic Therapy Tissue plasminogen activators have been administered to horses in an attempt to increase activity of plasmin and hence the rate of lysis of fibrin in the pleural cavity. Earlier attempts at fibrinolytic therapy used streptokinase or urokinase and were not beneficial. Use of modified compounds, such as alteplase and tenecteplase, is effective in hastening fibrinolysis, enhancing resolution of accumulated pleural fluid, and improving survival.6,9,10 There does not appear to be an increased risk of prolonged hemostasis. The procedure in one case involved intrapleural infusion of 12 mg of tenecteplase in 500 ml of isotonic saline after drainage of excessive pleural fluid.10 The treatment was repeated on three occasions over 10 days. Pharmacokinetics of alteplase in horses are described.11 A recommended protocol is infusion of tenecteplase (2–10 mg in 1–2 L of isotonic, polyionic fluid) q12 to 24h for 3 days, with a dwell time of 4 hours.6 Supportive Therapy Acutely or severely ill horses may be dehydrated and azotemic, and they may have acid-base disturbance. These horses should be treated with appropriate fluids administered intravenously. Pleuropneumonia is a painful disease, and every attempt should be made to relieve the horse’s chest pain. NSAIDs, including flunixin meglumine (1 mg/kg, orally, intramuscularly, or intravenously, every 8 hours)

995

or phenylbutazone (2.2 mg/kg, orally or intravenously, every 12 hours), often provide effective analgesia and presumably reduce inflammation in the pleural space. Horses should be provided with good nursing care, including a comfortable stall, free access to palatable water, and a good diet. Affected horses will often not eat adequately and should be tempted with fresh and nutritious fodder. Attention should be paid to the horse’s feet to detect early signs of laminitis and allow appropriate measures to be taken.

CONTROL

Prevention of pleuropneumonia involves reduction of risk factors associated with the disease. The main risk factors are other infectious respiratory diseases and transportation. Every effort should be made to prevent and treat respiratory disease in athletic horses, including institution of effective vaccination programs. Horses with infectious respiratory disease should not be vigorously exercised until signs of disease have resolved. Transportation of athletic horses is common and essential for their participation in competitive events. It cannot, therefore, be eliminated. Every effort should be made to minimize the adverse effects of transportation on airway health. Recommendations for transport of horses first made in 1917 are still relevant. Updated, these recommendations include the following: • Not transporting a horse unless it is healthy. Horses with fever should not be transported • Knowledgeable staff familiar with the horse should accompany it. • Suitable periods of rest and acclimation should be provided before recently transported or raced horses are transported. • The time during which horses are confined for transportation should be kept to a minimum. Horses should be loaded last and unloaded first in flights with mixed cargo. • The route taken should be the most direct and briefest available. • Horses should be permitted adequate time to rest at scheduled breaks. If possible, on long journeys horses should be unloaded and allowed exercise (walking) and access to hay and water. • Horses should have frequent, preferably continuous, access to feed and water during transportation. • Horses should not be exercised after arrival until they are free of fever, cough, or nasal discharge. • Horses should not be restrained during transportation such that they are unable or unwilling to lower their heads.

996

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

• Air quality should be optimal in the vehicle used to transport the horse. REFERENCES

1. Bodecek S, et al. Equine Vet Educ. 2011;23:296. 2. Hepworth-Warren KL, et al. Equine Vet Educ. 2015;27:283. 3. Ferrucci F, et al. Equine Vet Educ. 2008;20:526. 4. Tomlinson JE, et al. J Vet Int Med. 2015;n/a. 5. Rush BR, et al. Equine Vet Educ. 2011;23:302. 6. Tomlinson JE, et al. J Vet Int Med. 2015;n/a. 7. Belli CB, et al. Vet Rec. 2011;169. 8. Hilton H, et al. Vet Surg. 2010;39:847. 9. Hilton H, et al. Vet Rec. 2009;164:558. 10. Rendle DI, et al. Aust Vet J. 2012;90:358. 11. Baumer W, et al. BMC Vet Res. 2013;9.

ACUTE BRONCHO-INTERSTITIAL PNEUMONIA IN FOALS Acute broncho-interstitial pneumonia is a disease of foals less than 7 months of age, and usually less than 2 months of age, characterized by a rapid onset of respiratory distress. The condition is clinically similar to acute lung injury identified in other species.1 The etiology is unclear in many cases, but causes or agents associated with the disease include equine influenza virus infection,2 R. equi, equine herpesvirus-2, equine arteritis virus, or Pneumocystis carinii. The disease is likely a result of severe pulmonary injury by any of a number of infectious or toxic agents. The respiratory distress results from loss of pulmonary function because of necrosis of the epithelium of alveoli and terminal bronchioles. Foals typically present with an acute onset (80%) Thoroughbred and Standardbred racehorses, although clinical signs are less common. Occurs worldwide in any horse that performs strenuous exercise. Case-fatality rate is low, although because of the high incidence of the disease, deaths occur frequently during racing. Pathogenesis Probably associated with rupture of pulmonary capillaries by the high pulmonary vascular pressures generated during exercise. There does not appear to be a contributory role for preexisting inflammation and obstruction of

Treatment Furosemide is effective in decreasing the frequency and severity of the disease. Control There are no specific control measures; however, prevention of environmental and infectious respiratory disease might reduce the incidence of the disease.

ETIOLOGY Exercise-induced pulmonary hemorrhage of horses (EIPH) is a disease that occurs in horses during strenuous exercise.1 There is evidence of a genetic component to epistaxis in Thoroughbred racehorses (h2 = 0.27-0.5),2 but there are no reports of heritability or genetic factors contributing to EIPH.

EPIDEMIOLOGY

EIPH is primarily a disease of horses, although it occurs in racing camels and Greyhounds.3 EIPH occurs in horses worldwide, and there does not appear to be any geographic distribution. It is a disorder of horses that run at high speed, such as Thoroughbred or Standardbred racehorses. The disorder is uncommon in endurance horses and is rare in draft breeds, although it does occur in horses used for these activities.4 There is increasing recognition of its importance in sport horses (3-day event, show jumping, but not in dressage).5 The prevalence of EIPH varies with the method used to detect it and the frequency with which horses are examined, as discussed later in this section. Epistaxis associated with exercise is almost always attributable to pulmonary hemorrhage and occurs only in a small proportion of racehorses. Epistaxis occurs in only 3% of horses that have blood detected in the trachea by endoscopic examination performed within 2 hours of racing. The prevalence of epistaxis

998

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

in racehorses varies between 0.1% and 9.0%, with the frequency depending on the breed, age and sex of horses selected for study, the type of racing, and the timing and frequency of observation of horses after racing. Epistaxis is more common in older horses. There are conflicting reports of a sex predisposition, although epistaxis may be more common in female Thoroughbreds. Epistaxis is more common after races of less than 1600 m than in longer races, although not all sources agree on this point. However, horses in steeplechase races, which are typically longer than 2000 m, are at greater risk of epistaxis than are horses in flat races. Incidence of epistaxis in steeplechase horses in the United Kingdom is 5.3 per 1000 starts and 3.6 per 1000 starts in hurdle racing.6 Risk factors for horses in jumps races (steeplechase) include previous epistaxis (odds ratio [OR] 6.1 [4.4–8.3]), racing in a claiming race (OR 5.9 [1.4–25]), greater than 9 starts in previous 4 to 6 months (OR 10 [2–47]), and racing on firmer ground.6 Epistaxis is relatively uncommon, and most horses with EIPH do not have epistaxis. There are a variety of other methods of detecting EIPH, including endoscopic examination of the airways and microscopic examination of tracheal aspirates or bronchoalveolar lavage fluid. Almost all Thoroughbred racehorses in active training have hemosiderophages in bronchoalveolar lavage fluid, indicating that all have some degree of EIPH. The prevalence of EIPH decreases when diagnosis is based on endoscopic examination of horses after exercise or racing. EIPH is very common in Thoroughbred racehorses, with estimates of prevalence, based on a single endoscopic examination of the trachea and bronchi, of 43% to 75%.7,8 The prevalence increases with the frequency of examination, with over 80% of horses having evidence of EIPH on at least one occasion after examination after each of three consecutive races.9 There can be considerable variability in severity of EIPH within an individual horse on repeated examination over a racing season.8 The prevalence of EIPH in Standardbred racehorses is assumed to be lower, with 26% to 34% of horses reported to have blood in the trachea after racing. However, these studies were based on a single examination and one only reported as positive those horses with blood covering more than one half the tracheobronchial tree. When examined after each of three races, 87% of Standardbred racehorses have evidence of EIPH on at least one occasion, suggesting that EIPH is as common in Standardbred racehorses as it is in Thoroughbred racehorses. Exercise-induced pulmonary hemorrhage occurs in approximately 62% of racing Quarter Horses and has been observed in Quarter Horses used for barrel racing. The disorder occurs in racing Appaloosa horses.

Approximately 11% of polo ponies are affected with EIPH. The disease occurs in draft horses but is not well documented. Age is considered a risk factor for EIPH, with the prevalence of the disorder being higher in older horses, but the risk factor is the amount of racing that a horse has completed, not its age.10,11 There is no consistent association of sex with prevalence of EIPH. Among Thoroughbred racehorses there is an unclear relationship between the speed of racing and the risk of EIPH.10,12 Lesions of EIPH are not detected in young Thoroughbred racehorses that have trained at speeds of less than 7 m/s. The risk of EIPH increases with racing at lower ambient temperatures10,12 and with the wearing of bar shoes during racing.12 There is no association between risk of EIPH and track hardness.10,12

PATHOGENESIS

The cause of EIPH is rupture of alveolar capillary membranes with subsequent extravasation of blood into interstitial and alveolar spaces. The source of blood in such instances is the pulmonary circulation. Bleeding from bronchial circulation during exercise has been suggested, based on histologic evidence of bronchial angiogenesis in horses that have experienced previous episodes of EIPH, but contribution of the bronchial circulation to EIPH has not been demonstrated. Regardless of the contribution of bronchial circulation to blood in the airways, the likely initial lesion is in capillaries associated with the pulmonary circulation. There is increasing evidence that the primary lesion is arteriovenous remodeling of pulmonary veins.13-16 Remodeling of pulmonary veins results in loss of distensibility and partial occlusion to blood flow with subsequent presumed increases in pulmonary alveolar capillary pressure.13,17 Hemorrhage into the interstitial space and alveoli, with subsequent rostral movement of blood in the airways, results in blood in the trachea and bronchi. Rupture of alveolar capillaries occurs secondary to an exercise-induced increase in transmural pressure (pressure difference between the inside of the capillary and the alveolar lumen). If the transmural stress exceeds the tensile strength of the capillary wall, the capillary ruptures. The proximate cause of alveolar capillary rupture is the high transmural pressure generated by positive intracapillary pressures, which are largely attributable to capillary blood pressure, and the lower intraalveolar pressure generated by the negative pleural pressures associated with inspiration. During exercise, the absolute magnitudes of both pulmonary capillary pressure and alveolar pressure increase, with a consequent increase in transmural pressure. Strenuous exercise is associated with marked increases in pulmonary artery pressure in horses.

Values for mean pulmonary arterial pressure at rest of 20 to 25 mm Hg increase to more than 90 mm Hg during intense exercise because of the large cardiac output achieved by exercising horses. The increases in pulmonary artery pressure, combined with an increase in left atrial pressure during exercise, probably result in an increase in pulmonary capillary pressure. Combined with the increase in pulmonary capillary pressure is a marked decrease (more negative) in pleural, and therefore alveolar, pressure during exercise. The pleural pressure of normal horses during inspiration decreases from approximately –0.7 kPa (–5.3 mm Hg) at rest to as low as –8.5 kPa (64 mm Hg) during strenuous exercise. Together, the increase in pulmonary capillary pressure and decrease (more negative) in intrapleural (alveolar) pressure contribute to a marked increase in stress in the alveolar wall. Although the alveolar wall and pulmonary capillaries of horses are stronger than those of other species, rupture may occur because the wall stress in the alveolus exceeds the mechanical strength of the capillary. Other theories of the pathogenesis of EIPH include: small-airway disease, upper airway obstruction, hemostatic abnormalities, changes in blood viscosity and erythrocyte shape, intrathoracic sheer forces associated with gait, and bronchial artery angiogenesis. It is likely that the pathogenesis of EIPH involves several processes, including pulmonary hypertension, lower alveolar pressure, and changes in lung structure, that summate to induce stress failure of pulmonary capillaries. Obstruction of either the upper or lower airways has been proposed as a cause of EIPH. Inspiratory airway obstruction results in more negative intrapleural, and therefore alveolar, pressures. This effect is exacerbated by exercise, with the result that alveolar transmural pressure is greater in horses with airway obstruction. The higher transmural pressure in such horses may increase the severity of EIPH, although this has not been demonstrated. Moreover, although inspiratory airway obstruction may predispose to EIPH, the prevalence of this condition is much less than that of EIPH, indicating that it is not the sole factor inducing EIPH in most horses. Horses with moderate to severe EIPH have histologic evidence of inflammation of the small airways, and there is a clear association between the presence of EIPH and inflammatory changes in bronchoal­ veolar or tracheal aspirate fluid. However, instillation of autologous blood into the airways does not induce a marked inflammatory response in normal horses, and it is therefore unclear whether inflammation alone induces or predisposes to EIPH.18,19 Theoretically, small-airway inflammation and bronchoconstriction have the potential to produce intrathoracic airway obstruction

Diseases of the Equine Respiratory Tract

VetBooks.ir

and, therefore, a more negative alveolar pressure. Given that small-airway disease is common in horses, there is the potential for an important effect of factors such as viral infections, air pollution, and allergic airway disease to contribute to the initiation or propagation of EIPH. The characteristic location of lesions of EIPH in the caudodorsal lung fields has led to the proposal that hemorrhage is a result of tissue damage occurring when waves of stress, generated by forelimb foot strike, are focused and amplified into the narrowing cross-sectional area of the caudal lung lobes. According to the theory, the locomotor impact of the forelimbs results in transmission of forces through the scapula to the body wall, from where they pass into the lungs and caudally and dorsally. As the wave of pressure passes into the narrower caudodorsal regions of the lungs it generates progressively greater shearing forces that disrupt tissue and cause EIPH. However, studies of intrapleural pressures have not demonstrated the presence of a systemic pressure wave passing through the lung and do not provide support for this hypothesis. Horses with EIPH have been suspected of having defects in either hemostasis or fibrinolysis. However, although exercise induces substantial changes in blood coagulation and fibrinolysis, these is no evidence that horses with EIPH have defective coagulation or increased fibrinolysis. Regardless of the cause, rupture of pulmonary capillaries and subsequent hemorrhage into airways and interstitium causes inflammation of both airways and interstitium with subsequent development of fibrosis and alteration of tissue compliance. Heterogeneity of compliance within the lungs, and particularly at the junction of normal and diseased tissue, results in the development of abnormal shear stress with subsequent tissue damage. These changes are exacerbated by inflammation and obstruction of small airways, with resulting uneven inflation of the lungs. The structural abnormalities, combined with pulmonary hypertension and the large intrathoracic forces associated with respiration during strenuous exercise, cause repetitive damage at the boundary of normal and diseased tissue with further hemorrhage and inflammation. The process, once started, is lifelong and continues for as long as the horse continues to perform strenuous exercise.

CLINICAL FINDINGS

Poor athletic performance or epistaxis are the most common presenting complaints for horses with EIPH. Although poor performance may be attributable to any of a large number of causes, epistaxis associated with exercise is almost always secondary to EIPH. Epistaxis as a result of EIPH occurs during or shortly after exercise and is usually first noticed at the end of a race, particularly

999

when the horse is returned to the paddock or winner’s circle and is allowed to lower its head. It is usually bilateral and resolves within hours of the end of the race. Epistaxis may occur on more than one occasion, especially when horses are raced or exercised at high speed soon after an initial episode. Exercise-Induced Pulmonary Hemorrhage and Performance Failure of racehorses to perform to the expected standard (poor performance) is often, accurately or not, attributed to EIPH.7 Many horses with poor performance have cytologic evidence of EIPH on microscopic examination of tracheobronchial aspirates or bronchoalveolar lavage fluid or have blood evident on endoscopic examination of the tracheobronchial tree performed 30 to 90 minutes after strenuous exercise or racing. However, it is important to recognize that EIPH is very common in racehorses, and it should be considered the cause of poor performance only after other causes have been eliminated. Severe EIPH undoubtedly results in poor performance and, on rare occasions, death of Thoroughbred racehorses.1,7,20 Thoroughbred horses with EIPH have impaired performance compared with unaffected horses.7 Affected horses have a lower likelihood of finishing in the first three places, are less likely to be elite money earners, and finish further behind the winner than do unaffected horses. Results of studies in Standardbred racehorses indicate either a lack of effect of EIPH on performance or an association between EIPH and superior performance. There was no relationship between presence of EIPH and finishing position in 29 Standardbred racehorses with intermittent EIPH examined on at least two occasions, nor in 92 Standardbred racehorses examined on one occasion. However, of 965 Standardbred racehorses examined after racing, those finishing first or second were 1.4 times more likely (95% confidence interval 0.9-2.2) to have evidence of EIPH on tracheobronchoscopic examination than were horses that finished in seventh or eighth position. Physical Examination Apart from epistaxis in a small proportion of affected horses (Fig. 12-26), there are few abnormalities detectable on routine physical examination of horses with EIPH. Rectal temperature and heart and breathing rates may be elevated as a consequence of exercise in horses examined soon after exercise, but values of these variables in horses with EIPH at rest are not noticeably different from those of horses with no evidence of EIPH. Affected horses may swallow more frequently during recovery from exercise than do unaffected horses, probably as a result of blood in the larynx and pharynx. Coughing is common in horses recovering from strenuous exercise and after recovery from exercise; horses with

Fig. 12-26  Thoroughbred racehorse with epistaxis secondary to exercise-induced pulmonary hemorrhage during racing.

EIPH are no more likely to cough than are unaffected horses.1 Other clinical signs related to respiratory abnormalities are uncommon in horses with EIPH. Respiratory distress is rare in horses with EIPH and, when present, indicates severe hemorrhage or other serious lung disease such as pneumonia, pneumothorax or rupture of a pulmonary abscess. Lung sounds are abnormal in a small number of EIPH-affected horses and, when present, are characterized by increased intensity of normal breath sounds during rebreathing examination. Tracheal rales may be present in horses with EIPH but are also heard in unaffected horses. Tracheobronchoscopy Observation of blood in the trachea or large bronchi of horses 30 to 120 minutes after racing or strenuous exercise provides a definitive diagnosis of EIPH. The amount of blood in the large airways varies from a few small specks on the airway walls to a stream of blood occupying the ventral one-third of the trachea. Blood may also be present in the larynx and nasopharynx. If there is a strong suspicion of EIPH and blood is not present on a single examination conducted soon after exercise, the examination should be repeated in 60 to 90 minutes. Some horses with EIPH do not have blood present in the rostral airways immediately after exercise, but do so when examined 1 to 2 hours later. Blood is detectable by tracheobronchoscopic examination for 1 to 3 days in most horses, with some horses having blood detectable for up to 7 days. Bronchoscopic examination can be used to estimate the severity of EIPH through the use of a grading system. The interobserver repeatability of tracheobronchoscopic assessment of severity of EIPH using a grading scale of 0 to 4 is excellent, and this scoring system has been widely adopted for use (Fig. 12-27).7,8,12,21 • Grade 0: No blood detected in the pharynx, larynx, trachea, or mainstem bronchi. • Grade 1: Presence of one or more flecks of blood or two or more short

1000

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

of the biology of the disease or a management decision by owners and trainers.11 There is no association between measures of long-term performance measured over 10 years and grades 1, 2, or 3 of EIPH in Thoroughbred racehorses.11

A

B

C

D

Fig. 12-27  Grading of EIPH in Thoroughbred racehorses—Grades 1 (A), 2 (B), 3 (C), and 4 (D). (Reproduced with permission Hinchcliff et al. 2005.21)

(< one-quarter of the length of the trachea) narrow ( half the length of the trachea) or greater than 2 short streams occupying less than one-third of the tracheal circumference. • Grade 3: Multiple, distinct streams of blood covering more than one-third of the tracheal circumference. No blood pooling at the thoracic inlet. • Grade 4: Multiple, coalescing streams of blood covering greater than 90% of the tracheal surface with pooling of blood at the thoracic inlet. It is assumed that a higher score represents more severe hemorrhage, but although the repeatability of this scoring system has been established, the relationship between the amount of blood in the large airways and the actual amount of hemorrhage has not been established. Radiography Thoracic radiography is of limited use in detecting horses with EIPH. Radiographs

can demonstrate the presence of densities in the caudodorsal lung fields of some horses, but many affected horses have minimal to undetectable radiographic abnormalities.1 Examination of thoracic radiographs of horses with EIPH can be useful in ruling out the presence of another disease process, such as a pulmonary abscess, contributing to the horse’s pulmonary hemorrhage or poor athletic performance. Prognosis Horses that have experienced one episode of epistaxis are more likely to have a second episode. For this reason most racing jurisdictions do not permit horses with epistaxis to race for a period of weeks to months after the initial instance, with more prolonged enforced rest after a subsequent episode of epistaxis and retirement from racing after a third bout. The recurrence rate after one episode of epistaxis in Thoroughbred horses is approximately 13.5% despite affected horses not being permitted to race for 1 month after the initial episode. This high rate of recurrence suggests that the inciting pulmonary lesions have not healed. Long-term examination of performance of horses with EIPH indicates the horses with grade 4 EIPH have shorter racing careers, but it is not clear if this is a function

Clinical Pathology Examination of Airway Secretions   or Lavage Fluid The presence of red cells or macrophages containing either effete red cells or the breakdown products of hemoglobin (hemosiderophages) in tracheal or bronchoalveolar lavage fluid provides evidence of EIPH. Detection of red cells or hemosiderophages in tracheal aspirates or bronchoalveolar lavage fluid is believed to be both sensitive and specific in the diagnosis of EIPH. Examination of airway fluids indicates the presence of EIPH in a greater proportion of horses than does tracheobronchoscopic examination after strenuous exercise or racing. The greater sensitivity of examination of airway fluid is probably attributable to the ability of this examination to detect the presence of small amounts of blood or its residual products and the longevity of these products in the airways. Although endoscopic examination may detect blood in occasional horses up to 7 days after an episode of EIPH, cellular evidence of pulmonary hemorrhage persists for weeks after a single episode. Red blood cells and macrophages containing red cells are present in bronchoalveolar lavage fluid or tracheal aspirates for at least 1 week after strenuous exercise or instillation of autologous blood into airways, and hemosiderophages are present for at least 21 days and possibly longer. Recent studies have reported on the use of red cell numbers in bronchoalveolar lavage fluid as a quantitative indicator of EIPH. However, this indicator of EIPH severity has not been validated nor demonstrated to be more reliable or repeatable than tracheobronchoscopic examination and visual scoring. Furthermore, considerable concern exists over the suitability of red cell counts in bronchoalveolar lavage fluid for assessment of severity of EIPH given that an unknown area, although presumably small, of the lung is examined by lavage and that there is a risk that this area of lung may not be representative of the lung as a whole, similar to the situation of examination of bronchoalveolar lavage fluid of horses with pneumonia. Bronchoalveolar lavage of sections of both lungs, achieved using an endoscope, may obviate some of these concerns. Tracheal aspirates may be obtained any time after exercise by aspiration either during tracheobronchoscopic examination or through a percutaneous intratracheal needle. Aspirates obtained through an endoscope may not be sterile, depending on the collection technique. Bronchoalveolar lavage fluid

Diseases of the Equine Respiratory Tract

VetBooks.ir

can be obtained through either an endoscope wedged in the distal airway or a cuffed tube inserted blindly into a distal airway. Collection of fluid through an endoscope has the advantage of permitting examination of the distal airways and selection of the area of lung to be lavaged. However, it does require the use of an endoscope that is longer (2 m) than those readily available in most equine practices. Use of a commercial bronchoalveolar lavage catheter does not require use of an endoscope, and this procedure can be readily performed in field situations.

DIFFERENTIAL DIAGNOSIS Epistaxis and hemorrhage into airways can occur as a result of a number of diseases (Table 12-12).

Necropsy Exercise-induced pulmonary hemorrhage is a rare cause of death of racehorses, but among racehorses that die during racing for reasons other than musculoskeletal injuries, EIPH is common.20 Necropsy examination of horses is usually incidental to examination for another cause of death. Pertinent abnormalities in horses with EIPH are restricted to the respiratory tract. Grossly, horses examined within hours of strenuous exercise, such as horses examined because of catastrophic musculoskeletal injuries incurred during racing, may have severe petechiation in the caudodorsal lung fields. Horses with chronic disease have blue/gray or blue/brown discoloration of the visceral pleural surfaces of the caudodorsal lung fields that is often sharply demarcated, especially on the diaphragmatic surface. The discoloration affects both lungs equally, with 30% to 50% of the lung fields being discolored in severe cases. Affected areas do not collapse to the same extent as

1001

unaffected areas and, in the deflated lung, have a spleen-like consistency. On cut surface, the discolored areas of lung are predominantly contiguous with the dorsal pleural surface and extend ventrally into the lung parenchyma. Areas of affected lung may be separated by normal lung. There is proliferation of bronchial vessels, predominantly arteries and arterioles, in affected areas. Histologically, affected areas exhibit bronchiolitis, hemosiderophages in the alveolar lumen and interstitial spaces, and fibrosis of interlobular septa, pleura, and around vessels and bronchioles. Treatment Prevention of EIPH is contentious because it can involve the administration of medications on the day of racing. The efficacy of various interventions and medications has recently been evaluated in two systematic reviews, both of which concluded that there was moderately strong to strong evidence

Table 12-12  Causes of epistaxis in horses Disease

Epidemiology

Clinical signs and diagnosis

Hemorrhage into trachea or bronchi, sometimes with epistaxis Exercise-induced Horses after strenuous exercise. Epistaxis is a rare but very specific sign of EIPH. pulmonary hemorrhage Most common in Only occurs after exercise. (EIPH) Thoroughbred and Standardbred racehorses. Endoscopic examination of the airways is diagnostic,

Treatment and control Efficacy of various drugs used for treatment and control is debated.

Furosemide is used extensively before racing.

Trauma

Sporadic. Associated with trauma to head, neck, or chest.

Physical examination reveals site and nature of the trauma. Can require endoscopic examination of upper airways.

Symptomatic treatment.

Pneumonia

Recent transport or respiratory disease. Can occur as outbreaks though usually individual animals.

Fever, tachypnea, abnormal lung sounds, leukocytosis; radiography demonstrates lung lesions. Cytologic and microbiological examination of tracheal aspirate.

Antimicrobials, NSAIDs, oxygen. Control by vaccination and prevention of respiratory disease.

Lung abscess

Sporadic. Hemorrhage can occur after exercise.

Sometimes no premonitory signs. Fever, depression, anorexia, cough. Hemogram demonstrates leukocytosis. Hyperfibrinogenemia. Ultrasonography or radiography demonstrates lesion. Tracheal aspirates.

Antibiotics.

Intrabronchial foreign body

Sporadic.

Cough, hemoptysis, fever. Endoscopy or radiography reveals foreign body.

Removal of foreign body—often not readily achieved.

Pulmonary neoplasia

Sporadic. Often older horse, but not always. Hemangiosarcoma.

Cough, hemoptysis. Demonstrate mass on ultrasonographic or radiographic examination.

None.

Severe, life-threatening epistaxis. Tachycardia, anemia, hemorrhagic shock.

Surgical ligation or occlusion of arteries in the guttural pouch.

Epistaxis not associated with exercise.

Surgery or injection of mass

Usually unilateral.

with formaldehyde.

Epistaxis (in addition to the previously listed diseases) Guttural pouch mycosis Sporadic. Acute-onset epistaxis. Ethmoidal hematoma

Sporadic.

Thrombocytopenia

Sporadic.

Epistaxis, mild, intermittent. Petechiation and ecchymotic hemorrhages. Thrombocytopenia.

Glucocorticoids.

Neoplasia

Sporadic.

Neoplasia of upper airways.

None.

Trauma

Sporadic.

Injury to head or pharynx.

Symptomatic.

Sinusitis

Sporadic.

Endoscopic or radiographic examination of sinus.

Drainage. Antimicrobials.

1002

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

that administration of furosemide before racing reduces the frequency and severity of EIPH in Thoroughbred racehorses.1,22 There was either weak evidence or no evidence of efficacy of other interventions. There is a recommendation for use of furosemide, but because of the regulatory issues related to its use, this is only a weak recommendation.1 Therapy of EIPH is usually a combination of attempts to reduce the severity of subsequent hemorrhage and efforts to minimize the effect of recent hemorrhage. Treatment of EIPH is problematic for a number of reasons. First, the pathogenesis of EIPH has not been determined although the available evidence supports a role for stress failure of pulmonary capillaries secondary to exerciseinduced pulmonary hypertension. Second, there is a lack of information using large numbers of horses under field conditions that demonstrates an effect of any medication or management practice (with the exception of bedding) on EIPH. There are numerous studies of small numbers of horses (< 40) under experimental conditions but these studies often lacked the statistical power to detect treatment effects and, furthermore, the relevance of studies conducted on a treadmill to horses racing competitively is questionable.1 Treatments for EIPH are usually intended to address a specific aspect of the pathogenesis of the disease and will be discussed in that context but should be considered in the context of the amount and strength of evidence, which for most treatments is scant and weak. Prevention of Stress Failure of the Pulmonary Capillaries There is interest in reducing the pressure difference across the pulmonary capillary membrane in an effort to reduce EIPH. Theoretically, this can be achieved by reducing the pressure within the capillary or increasing (making less negative) the pressure within the intrathoracic airways and alveolus. Reducing Pulmonary Capillary Pressure Furosemide administration as prophylaxis of EIPH is permitted in a number of racing jurisdictions worldwide, most notably Canada, the United States, Mexico, and most of the South American countries. Within the United States and Canada, almost all Thoroughbred, Standardbred, and Quarter Horse racing jurisdictions permit administration of furosemide before racing. The efficacy of furosemide in treatment of EIPH is now well documented.9,22 The mechanism by which furosemide reduces the severity of EIPH is unknown, although it is speculated that furosemide, by attenuating the exercise-induced increase in pulmonary artery and pulmonary capillary pressure of horses, reduces the frequency or severity of pulmonary capillary rupture.

Furosemide is associated with superior performance in both Thoroughbred and Standardbred racehorses, which further complicates assessment of its efficacy in treating EIPH. An increase in pulmonary capillary pressure secondary to altered rheostatic properties of blood during exercise has been suggested as a possible contributing factor for EIPH. Increasing Alveolar Inspiratory Pressure Airway obstruction, either intrathoracic or extrathoracic, increases airway resistance and results in a more negative intrathoracic (pleural) pressure during inspiration to maintain tidal volume and alveolar ventilation. Causes of extrathoracic airway obstruction include laryngeal hemiplegia and other abnormalities of the upper airway, whereas intrathoracic obstruction is usually a result of bronchoconstriction and inflammatory airway disease. Horses with partial extrathoracic inspiratory obstruction or bronchoconstriction and airway inflammation associated with recurrent airway obstructive disease (heaves) have pleural (and hence alveolar) pressures that are lower (more negative) than those in unaffected horses or in horses after effective treatment. Hypothetical relationships between the horse’s bit, airway obstruction and EIPH are not supported to date by empirical evidence.23,24 Partial inspiratory obstruction, such as produced by laryngeal hemiplegia, exacerbates the exercise-induced decrease in intrapleural pressures with a consequent increase in transmural capillary pressures. These changes may exacerbate the severity of EIPH, although an association between upper airway obstructive disease and EIPH has not been demonstrated. Surgical correction of airway obstruction is expected to resolve the more negative intrapleural pressure, but its effect on EIPH is unknown. Recently the role of the nares in contributing to upper airway resistance, and hence lowering inspiratory intrapleural pressure during intense exercise, has attracted the attention of some investigators. Application of nasal dilator bands (Flair strips) reduces nasal resistance by dilating the nasal valve and reduces red cell count of bronchoalveolar lavage fluid collected from horses after intense exercise on a treadmill. Furthermore, application of the nasal dilator strips to horses in simulated races reduces red cell count in bronchoalveolar lavage fluid of some, but not all, horses. The role of small-airway inflammation and bronchoconstriction in the pathogenesis of EIPH is unclear. However, horses with EIPH are often treated with drugs intended to decrease lower airway inflammation and relieve bronchoconstriction. Beta-adrenergic bronchodilatory drugs such as clenbuterol and albuterol (salbutamol) are effective in

inducing bronchodilation in horses with bronchoconstriction, but their efficacy in preventing EIPH is either unknown or, in very small studies, is not evident. Cortico­ steroids, including dexamethasone, fluticasone, and beclomethasone, administered by inhalation, parenterally, or enterally, reduce airway inflammation and obstruction but have no demonstrated efficacy in preventing EIPH. Cromolyn sodium (sodium cromoglycate) has no efficacy in preventing EIPH. Water vapor treatment (inhalation of water-saturated air) has been proposed as a treatment for EIPH because of its putative effect on small-airway disease. However, water vapor treatment has no effect on EIPH. The use of bedding of low allergenic potential (shredded paper) to prevent EIPH has no apparent effect on prevalence of the condition. Although it is suggested that preventing or minimizing small-airway disease may reduce the severity of EIPH, studies to demonstrate such an effect have not been reported. However, optimizing the air quality in barns and stables and preventing infectious respiratory disease appear to be sensible precautions. Interstitial Inflammation and Bronchial Angiogenesis Hemorrhage into interstitial tissues induces inflammation with subsequent development of fibrosis and bronchial artery angiogenesis. The role of these changes in perpetuating EIPH in horses is unclear but is probably of some importance. Treatments to reduce inflammation and promote healing with minimal fibrosis have been proposed. Rest is an obvious recommendation, and many racing jurisdictions have rules regarding enforced rest for horses with epistaxis. Although the recommendation for rest is intuitive, there is no information that rest reduces the severity or incidence of EIPH in horses with prior evidence of this disorder. Similarly, corticosteroids are often administered, either by inhalation, enterally or parenterally, in an attempt to reduce pulmonary inflammation and minimize fibrosis. Again, the efficacy of this intervention in preventing or minimizing the severity of EIPH has not been documented. Excessive Bleeding Coagulopathy and Fibrinolysis Exercise induces substantial changes in blood coagulation and fibrinolysis. However, there is no evidence that horses with EIPH have defective coagulation or increased fibrinolysis. Regardless, aminocaproic acid, a potent inhibitor of fibrin degradation, has been administered to horses to prevent EIPH. The efficacy of aminocaproic acid in preventing EIPH has not been demonstrated.1 Similarly, estrogens are given to horses with the expectation of improving hemostasis, although the effect of estrogens on coagulation in any species is unclear.

Diseases of the Equine Respiratory Tract

VetBooks.ir

There is no evidence that estrogens prevent EIPH in horses. Vitamin K is administered to horses with EIPH, presumably in the expectation that it will decrease coagulation times. However, because EIPH is not associated with prolonged bleeding times, it is unlikely that this intervention will affect the prevalence or severity of EIPH. Platelet Function Aspirin inhibits platelet aggregation in horses and increases bleeding time. Seemingly paradoxically, aspirin is sometimes administered to horses with EIPH because of concerns that increased platelet aggregation contributes to EIPH. There is no evidence that aspirin either exacerbates or prevents EIPH. Capillary Integrity Capillary fragility increases the risk of hemorrhage in many species. Various bioflavonoids have been suggested to increase capillary integrity and prevent bleeding. However, hesperidin and citrus bioflavonoids have no efficacy in prevention of EIPH in horses. Similarly, vitamin C is administered to horses with EIPH without scientific evidence of any beneficial effect. Summary of Treatment Options Selection of therapy for horses with EIPH is problematic. Given that most horses have some degree of pulmonary hemorrhage during most bouts of intense exercise, the decision must be made not only as to the type of treatment and its timing but also which horses to treat. Moreover, the apparently progressive nature of the disease with continued work highlights the importance of early and effective prophylaxis and emphasizes the need for studies of factors such as air quality and respiratory infections in inciting the disorder. The currently favored treatment for EIPH is administration of furosemide before intense exercise. Its use is permitted in racehorses in a number of countries but is contentious in many. A frequent practice is to administer furosemide before high-speed training, and not on the day of racing, in jurisdictions that do not permit race day administration of medications. There is increasing interest in the effect of furosemide administered 24 hours before racing; its efficacy in this situation remains to be determined. The association between furosemide administration and superior performance in Standardbred and Thoroughbred racehorses should be borne in mind when recommending use of this drug. Prevention and Control There are no documented preventive strategies. Rest is an obvious recommendation for horses with EIPH, but the hemorrhage is likely to recur when the horse is next

strenuously exercised. The duration of rest and the optimal exercise program to return horses to racing after EIPH is unknown, although some jurisdictions require exercise no more intense than trotting for 2 months. Firm recommendations cannot be made on duration of rest because of a lack of objective information. Although a role for lower airway disease (either infectious or allergic) in the genesis of EIPH has not been demonstrated, control of infectious diseases, and minimization of noninfectious lower airway inflammation appears prudent. Concern about the role of impact waves in the genesis of EIPH has led to discussion of “low-stress” training protocols, but these have not been adequately evaluated. FURTHER READING Hinchcliff KW, et al. Exercise-induced pulmonary hemorrhage: American College of Veterinary Internal Medicine consensus statement. J Vet Intern Med. 2015;29:743-758. Sullivan SL, et al. A systematic review and meta-analysis of the efficacy of furosemide for exercise-induced pulmonary haemorrhage in Thoroughbred and Standardbred race horses. Equine Vet J. 2015;47:341-349.

REFERENCES

1. Hinchcliff KW, et al. J Vet Int Med. 2015;29:743. 2. Velie BD, et al. Vet J. 2014;202:274. 3. Epp TS, et al. Comp Exerc Physiol. 2008;5:21. 4. Sullivan S, et al. Vet Clin Equine. 2015;31:187. 5. Van Erck-Westergren E, et al. Equine Vet J. 2013;45:376. 6. Reardon RJM, et al. Vet J. 2015;205:44. 7. Morley PS, et al. Equine Vet J. 2015;47:358. 8. Preston SA, et al. Equine Vet J. 2015;47:366. 9. Hinchcliff KW, et al. JAVMA. 2009;235:76. 10. Hinchcliff KW, et al. Equine Vet J. 2010;42:228. 11. Sullivan SL, et al. Equine Vet J. 2015;47:350. 12. Crispe EJ, et al. Equine Vet J. 2015;n/a. 13. Derksen F, et al. Compendium (Yardley, PA). 2011;33:E6. 14. Derksen FJ, et al. Equine Vet J. 2009;41:586. 15. Stack A, et al. Am J Vet Res. 2013;74:1231. 16. Stack A, et al. J Appl Phys. 2014;117:370. 17. Williams KJ, et al. Vet Pathol. 2008;45:316. 18. Derksen FJ, et al. Equine Vet J. 2007;39:334. 19. Williams KJ, et al. Equine Vet J. 2011;43:354. 20. Lyle CH, et al. Equine Vet J. 2012;44:459. 21. Hinchcliff KW, et al. Am J Vet Res. 2005;66:596. 22. Sullivan SL, et al. Equine Vet J. 2015;47:341. 23. Cook WR. Equine Vet Educ. 2014;26:381. 24. Cook WR. Equine Vet J. 2014;46:256.

RECURRENT AIRWAY OBSTRUCTION (HEAVES) SYNOPSIS Etiology Combined genetic predisposition with environmental challenge of inhaled barn and feed dust containing inciting agents that can include particles of molds, endotoxin, mites, plant debris, and inorganic material. Epidemiology Predominantly a disease of horses stabled in poorly ventilated barns

1003

and fed quality hay. Occurs worldwide but more commonly in the northern hemisphere. Increased prevalence in older horses. No breed or sex predilection. Clinical signs Range of severity of clinical signs. Chronic cough, mucopurulent nasal discharge, poor athletic performance, increased respiratory rate, increased expiratory effort, wheezes on thoracic auscultation, and abundant mucopurulent material in the trachea on endoscopic examination. Clinical pathology Neutrophilia in tracheal aspirate and bronchoalveolar lavage fluid. Lesions Bronchiolitis with mononuclear cell infiltration, epithelial, and goblet cell hyperplasia, neutrophil accumulation in airway lumens, and alveolar hyperinflation. Diagnostic confirmation Clinical signs, examination of bronchoalveolar lavage fluid, and the response to treatment. Treatment Remove the inciting cause by providing a dust-free environment, and administer corticosteroids. Bronchodilators are useful for treatment of acute bronchoconstriction. Control Prevent exposure to inciting cause. Ensure optimal air quality in stables or maintain horses at pasture.

Recurrent airway obstruction (RAO; heaves) is a recurrent disease of stabled adult horses characterized by neutrophilic airway inflammation and airway obstruction manifest clinically by the presence of coughing, excess mucus accumulation in airways, neutrophilic bronchoalveolar lavage fluid or tracheal aspirate, bronchospasm, tachypnea and increased respiratory effort, and exercise intolerance. Clinical severity ranges from mild coughing with minimal exercise intolerance during infrequent recurrences of the disease through to severe and persistent coughing, airway obstruction, and markedly increased work of breathing and abnormal breathing pattern. Removal of exposure to hay and straw and keeping the horse at pasture results in remission of the disease. The disease should be differentiated from the usually transient inflammatory airway disease of young adult horses in which there is no clinically significant impairment of pulmonary function. The disease is classically considered to have an allergic component.

ETIOLOGY Genetics There is a genetic component to the disease, although the precise pattern of inheritance and gene association is yet to be determined.1 There is a familial pattern to the disease in some breeds, and for Warmbloods and Lipizzan, affected parents increase the likelihood of off-spring developing recurrent airway obstruction (RAO) by fivefold.2 Segregation analysis indicates a mixed mode of

1004

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

inheritance in Warmbloods.3 It appears likely that multiple genes are involved in the predisposition to RAO and the genes involved might differ with breed of horse or other animal or environmental factors. Evidence for a multigene cause for predisposition to RAO includes the finding of quantitative trait loci (QTL) located on two different chromosomes (ECA 13 and ECA 15) in different families of affected Warmbloods4,5 and failure to identify a monogenetic predisposition to RAO. Further studies in half-sibling horses and from unrelated horses indicates that at least one causative variant is a QTL region located on ECA 13 that is not associated with any coding variants, suggesting that the cause is a regulatory mutation.6 The cause does not appear to be abnormalities in the gene encoding DNAH3 (dynein—a component of cilia), although it is located in this QTL and has 53 polymorphisms including 7 nonsynonymous variants.7 Similarly, mutations in the integrin alpha X gene, which is related to allergy in humans and is located in the region of the QTL in ECA 13, are not associated with RAO.4,8 However, expression of interleukin 4 receptor gene, which is also located in the region of the QTL, is greater in bronchoalveolar lavage fluid of RAOaffected half-siblings than in unaffected animals, suggesting a role for this cytokine, or a mutation in its gene, in RAO.9 Environmental Factors RAO is caused by inhalation by susceptible horses of dust particles found in barns, bedding, and feed materials such as dusty hay. The inhaled particles include endotoxin, mites, plant debris, inorganic materials, and conidia and fragments of molds. Faenia rectivirgula (formerly known as Micropolyspora faeni), Aspergillus fumigatus, and Thermoactinomyces vulgaris are molds commonly associated with respiratory disease in susceptible horses, as evidenced by experimental studies involving inhalation of mold or mold fragments by horses. Molds contain a number of inflammatory substances, including various allergens, glucans, mycotoxins, and proteases, and it is not clear which of these agents are the inciting cause of RAO. Furthermore, dust containing mold also contains endotoxin. Inhalation of fungal spores, endotoxin and silica microspheres causes RAO in susceptible horses but not in healthy horses.10 There was neutrophilic inflammation in both healthy and RAO horses but bronchospasm in only RAO horses.10 Endotoxin contamination of molds contributes to the airway response to inhalation of preparations of molds used in experimental studies and inhalation of endotoxin alone produces airway inflammation and impaired respiratory function in horses in a dose-dependent manner, with RAO-susceptible horses having an exaggerated response at lower doses. Endotoxin concentrations in the breathing zone of horses are eight times higher in

stables than at pasture.11 However, the response to endotoxin is less than that of susceptible horses exposed to hay dust containing endotoxin, indicating that endotoxin alone is not sufficient to cause the clinical signs of RAO. Other compounds in hay dust are integral to the development of RAO. It is emphasized that there is not one causative agent acting alone but rather a range of agents that, when inhaled in sufficient concentration by susceptible horses, induce airway disease. It is likely that RAO is associated with the potentiating interactions among several agents present in barn or hay dust and is not simply a response to one agent. The mechanisms underlying development of airway inflammation and respiratory dysfunction are provided under “Pathogenesis.” Viral infections and 3-methylindole intoxication are not considered important causes of RAO.

EPIDEMIOLOGY Occurrence Although RAO is one of the more common diseases of horses and is a major cause of loss of performance and wastage in European horses, there are few reports of its epidemiologic characteristics. The disease is common in Europe and North America but is rare in Australia. The prevalence of RAO in the Great Britain, based on a random survey of owners who use veterinary surgeons, is 14% (95% confidence interval of 10.7%–17.4%).12 The 7-day incidence (ie, new occurrence of the disease in an animal) of RAO in horses and ponies in Great Britain is 0.4% (0%– 0.8%), and the prevalence of RAO was 5.8% (95% CI 4.2%–7.5%), as reported by owners.13 In Germany, 83% of horses believed to be healthy at an auction were found to have clinical evidence of chronic pulmonary disease. Inflammatory airway disease is very common in horses, with 96% of racehorses in Hong Kong examined at necropsy and 27% of healthy racehorses in training having an increased proportion (>20%) of neutrophils in tracheal aspirate, indicating inflammatory airway disease. Among stabled pleasure horses in Michigan, ~17% had cytologic or endoscopic evidence of airway inflammation,11 and 12% of horses examined in an abattoir in the northern United States had histologic evidence of bronchitis. However, although airway inflammation is a component of RAO, the airway inflammation common in young athletic horses and stabled horses is not generally considered to be RAO or necessarily a prodrome of RAO. The case-fatality rate for moderately to severely affected horses is approximately 20% over a 2- to 4-year period. Among horses greater than 15 years of age, presence of RAO is not significantly associated with death (P = 0.73, hazard ratio 1.19, 95% CI 0.4–3.2).14 Most mildly to moderately affected

horses respond well to treatment and continue to perform at a satisfactory level. Risk Factors Animal Risk Factors The disease occurs in adult horses and ponies. A survey of horse and pony owners in Great Britain found that the median age of horses and ponies with the disease is 18.2 years versus 12.7 for unaffected animals.13 Another owner survey found the median age of RAO affected horses as 13 years (interquartile range [IQR] of 9.5–20 years), whereas that of unaffected horses was 10 years (IQR 7–14.4 y).12 The odds of a horse having RAO (as reported by the owner) increases with greater age (odds ratio of 5.1, 8.1, 11.4, 9.5, and 18.3 for horses 5–7, 7–9, 9–11, 11–15, and greater than15 years of age compared with horses < 5 years).12 Examination of a convenience sample stratified random sample of 3000 horses of 1e to 40 years of age in the Netherlands revealed spontaneous coughing during a 10 minute observation period in 1% of horses, nasal discharge in 1.9%, and abnormal respiratory effort in 1%.15 Of 200 horses and ponies greater than 15 years of age in the United Kingdom randomly chosen for examination by a veterinarian, 13.6% had marked abnormalities (expiratory wheeze, cough and/or increased abdominal effort) during rebreathing examination that are consistent with RAO.16 A further 17.8% had moderately severe abnormalities. Those horses and ponies with abnormalities identified during rebreathing examination were significantly older (median 21.2 years) than were animals without abnormalities (18.0 years).16 Similarly, 15% of horses greater than 30 years of age have marked clinical signs consistent with RAO, and a further 19% have moderate abnormalities.17 There is no apparent breed, sex, or height predisposition,12 with the exception that Thoroughbreds are 3 times more likely to be examined for the disease than are ponies, although this could represent a sampling bias in that owners of Thoroughbreds might be more likely to seek veterinary attention than owners of ponies. The finding of increased likelihood of Thoroughbred horses having the disease is not consistent among studies. A survey of donkey owners in the United Kingdom did not elicit any reports of signs consistent with RAO in any of the ~1700 animals.18 This might represent underreporting of the condition in donkeys or a low prevalence of the disease in donkeys. There are horses that develop the disease and other horses, maintained in an identical situation, that do not.12 Development of disease is dependent on the horse being susceptible to the inflammatory effect of inhaled dust but the reasons for this individual susceptibility are poorly understood. As noted earlier (“Etiology—Genes”), familial predisposition has been suggested based on the observation that Lipizaners and German and

Diseases of the Equine Respiratory Tract

Environmental Risk Factors Season Horses are approximately 2 times more likely to be examined by a veterinarian because of the disease in winter or spring compared with summer, suggesting a seasonality to the occurrence of the disease perhaps as a result of increased stabling during winter. Signs of respiratory disease in horses with RAO are ~2 times more likely to occur in winter months, with peak values of 45% to 50% of RAO-affected horses having clinical signs of the disease in January and February in Great Britain (Fig. 12-28). Housing and Hay Feeding There is a clear association between housing, feeding of hay, and development of the disease. Typically, susceptible horses are clinically normal when at pasture and develop signs of disease within hours to days of being housed in stables and fed dusty hay. Moving affected horses to pasture, or improving air quality by increasing ventilation and feeding processed feedstuffs, results in resolution of the disease. Horses living in urbanized environments are approximately twice as likely to have the disease. Although the reason for this association has not been demonstrated, it is reasonable to assume that at least part of the increased risk is attributable to poorer air quality for horses in an urban environment.12 Management practices that might contribute to development or exacerbation of RAO vary widely around the world, with differing practices related to duration of stabling, type of bedding, air quality in stables

and such. Within Great Britain, 4% of horses are stabled 24 hours per day year round and 9% stabled all day (24 hours) in winter.19 61% are stabled part of each day with pasture turn-out and 36% are turned for 24 hours each day.19 Development of disease is related to inhalation of respirable particles that gain access to the lower respiratory tract. Respirable particles are less than 5 µm diameter, the principal source of these particles in stalls is hay, and the majority of particles are fungal spores. The concentration of particles in air of the stable is determined by the rate of release of particles from hay, which is dependent in large part on the quality of the hay, concentration of fungal spores in the hay, and the rate of clearance of dust from the stable, a function of the ventilation rate. Concentrations of respirable dust particles in the breathing zone of stabled horses can be as high as 20 mg/m3. The severity of increases in neutrophil count and proportion and decreases in pulmonary function in experimental models of RAO are related in a dosedependent fashion to the amount of dust inhaled. The presence of dust particles, and not the soluble products in hay dust, is responsible for most of the airway neutrophilia induced by inhalation of hay dust. Hay is the usual original source of spores in stable air. However, decomposing wood shavings are also a source of spores of fungi that multiply during degradation of plantbased materials, and housing horses in poorly ventilated stalls deeply bedded with wood shavings may be detrimental to their respiratory health. Spores from hay enter the bedding either directly or after dispersal through the air and multiply in the bedding if it is not removed regularly. Diced paper and wood shavings, when fresh, usually contain very few spores. Barley and wheat straw are usually free of any small spores such as A. fumigatus or M. faeni. Bedding horses on fresh wood shavings and feeding a nutritionally complete pelleted ration results in a respirable dust burden 3% of that of horses fed hay and bedded with straw. Dust burdens measured in the air of the stall underestimate the respirable particle challenge of horses

50 40 30 20 10 December

November

October

September

August

July

June

May

April

March

February

0 January

Relative frequency (%)

VetBooks.ir

Swiss Warmbloods are 3.2 times more likely to have RAO if one parent was affected and 4.6 times as likely if both parents had RAO. There is no association between major histocompatibility markers (equine leukocyte antigens) and occurrence of RAO. Exposure to inciting agents is associated with a variety of environmental factors, including potentially outdoor concentrations of aeroallergens and climatic factors but most importantly housing and feeding practices.

Fig. 12-28  Proportion of horses in Great Britain with recurrent airway obstruction (RAO) that display clinical signs of respiratory disease in each month. (Reproduced with permission.12)

1005

because of the high concentration of particles in hay and bedding, areas from which the horse inhales while eating. Respiratory health of horses is related to stable design and ventilation, with horses in poorly ventilated barns having more respiratory disease than horses in well-ventilated barns. See “Control” for recommendations regarding stable design.

PATHOGENESIS

Susceptible horses, when exposed to adequate concentrations of respirable dust in the breathing zone, develop airway inflammation including neutrophilia in airway secretions, excess mucus accumulation, and bronchospasm within hours to days of exposure. Longer-term changes include bron­ chiolitis with peribronchial lymphocytic infiltration and increased thickness of submucosal smooth muscle and bronchial epithelium. Notably, eosinophils are not an important component of the inflammatory response in horses with RAO, either in bronchoalveolar lavage fluid or in peribronchial infiltrates.20 These morphologic changes contribute to the reduction in airway diameter that underpins the physiologic effect of the disease. Emphysema and bronchiectasis develop as the severity of the disease worsens. The mechanisms underlying these responses to inhalation of dust are not well defined but can be considered in the contexts of immune and inflammatory responses, mucus secretion, and pulmonary dysfunction. Inflammatory and Immune Responses The precise immunologic abnormalities and mechanisms causing airway and peribronchial inflammation in affected horses is unclear.21 Inflammation is associated with excessive mucus production, airway swelling, and abnormal lung function. The inflammatory response in horses with RAO is neutrophilic, with lesser numbers of mast cells and rarely eosinophils.20 The mechanisms underlying this inflammatory response have not been fully elucidated, but differing responses in RAO and healthy horses, and differing responses among families of RAO sensitive horses, to antigenic challenge supports an acquired immune-mediated process.22 Furthermore, upregulated expression of IL1β, IL8, TLR4, TNFα, TGFβ1, and NFkβ transcripts in RAO-affected compared with healthy horses and the strong correlation with clinical variables indicative of disease severity provide support for an immune-mediated disease process.23 There is currently no clear consensus on the immune mechanisms involved in development or perpetuation of RAO despite numerous studies examining cell types,20,24-28 cytokines,23,29-34 gene expression,23,29,31,35 and antibody isotypes.36 The presence of allergen-specific IgE antibodies in bronchoalveolar lavage fluid is supportive of a hypersensitivity reaction (type 1

1006

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

hypersensitivity response), as is the observation that reaginic antibodies (IgE and some classes of IgG) in serum of horses sensitive to Aspergillus fumigatus cause mast cell (rat origin) degranulation.36 Others have proposed type 3 and type 4 immune reactions as the basis of the disease, and there are suggestions of polarized Th1 or Th2 responses. Detection of increased mRNA expression of thymic stromal lymphopoietin, a cytokine involved in lymphocyte development and driving a Th2 response, in bronchoalveolar lavage fluid (BALF) and peribronchial cells of horses with RAO is evidence of a con­ tribution of a Th2 mechanism to RAO.32 One proposed explanation is that RAOsusceptible horses exhibit a Th2-like immune response to inhalation of hay or barn dust characterized by increased expression of interleukins 4 and 5 and decreased expression of interferon-γ in cells obtained by bronchoalveolar lavage. Others have not detected a pure Th2-like cytokine profile, finding instead a mixed inflammatory response including increases in expression (mRNA) in cells obtained from bronchoalveolar lavage fluid of affected horses of interferon-γ, tumor necrosis factor-α, interleukins 1β and 4, and interleukins 8 and 17 (potent attractors of neutrophils) but not interleukins 2, 5, and 10. However, all the studies cited previously were performed on crude preparations of cells obtained by bronchoalveolar lavage, and the results could have been influenced by the varying proportions of types of cells in these preparations. A study examining just CD4 and CD8 lymphocytes in blood and bronchoalveolar lavage fluid of RAO-affected horses demonstrated a general down-regulation in expression of interferon-γ, and interleukins 4, 5, and 13 and no evidence of a cytokine profile consistent with either sole or predominate Th1- or Th2-like responses. The magnitude of the inflammatory response varies depending on the challenge (i.e., nature of the inhaled material) with responses to endotoxin characteristically being less than that of hay dust. Regardless of the underlying mechanism, it is clear that T cells are involved in mediating and likely modulating the response to exposure to inciting agents in susceptible horses25 and this results in airway inflammation and interference with normal respiratory function. The role of the observed increased rate of T-cell apoptosis in resolution of clinical signs after removal of the inciting agents in RAO-affected horses is uncertain.27 Following inhalation of inciting agents there is recruitment of neutrophils, but not eosinophils or platelets, into the lungs in most horses that develop changes in lung function. Histologically, there is peribronchiolar accumulation of lymphocytes and luminal accumulations of neutrophils in affected horses. The neutrophilic response is airways is mediated at least in part by IL-8 and IL-17 and by MAPK and PI3K pathways

in horses with heaves.30 The entry of neutrophils into the airways is associated with activation of neutrophils and platelets. Neutrophil activation occurs in horses with RAO 10 and 24 hours after antigen challenge and is mediated by increased expression of CD13 on circulating neutrophils of susceptible horses exposed to inciting dust.37 However, there is not increased expression of mRNA for TNF-alpha, IL-beta, IL-8, and MIP-2 in horses with RAO, suggesting that release of these cytokines is not necessary for the neutrophilic response characteristic of the disease.38 The neutrophils of horses during episodes of RAO, but not when the horses are asymptomatic, have increased adherence in vitro to protein coated plastic suggesting a mechanism for the increased migration of neutrophils into airways of affected horses. However, neutrophils of asymptomatic RAO-affected horses (ie, those horses in remission of the disease) have an exaggerated serum concentration of TNF and mRNA expression after lipopolysaccharide exposure compared with cells of healthy horses.26 A greater proportion of neutrophils in BALF of RAO-affected horses are viable and have a slower rate of apoptosis compared with those of unaffected horses, suggesting a role for increased neutrophil survival in airways in horses with RAO.28 Inhibition of neutrophil phosphodiesterase-4 activity does not alleviate clinical signs of RAO or decrease neutrophil numbers in bronchoalveolar lavage fluid in affected horses, suggesting that neutrophils are not primarily involved in the genesis of airway obstruction. The extent to which neutrophils in the airways are activated has not been determined, and their role in the development of respiratory dysfunction is unclear given that glucocorticoid administration attenuates the respiratory dysfunction but not airway neutrophilia in horses with RAO (see under “Treatment”). Airway inflammation is associated with increases in concentration of inflammatory mediators including leukotriene B4, prostanoids including thromboxane, and proteases. Activity of matrix metalloproteinase-9 is higher in horses with RAO than in unaffected horses and is induced in a dosedependent manner by inhalation of inciting substances including hay dust and endotoxin. MMP-9 is likely important in the inflammatory process associated with RAO through excessive gelatinolytic proteolysis that can contribute to lung injury and through a role in lung remodeling. Inflammation is also associated with increased oxidative stress in lungs of horses with RAO as indicated by elevated concentrations of epiPGF2a and redox ratio of glutathione in pulmonary lavage fluid. RAO is associated with platelet activation and increases in mean platelet volume, indicating consumption of platelets and bone marrow release of younger throm­bocytes.35,37

Platelets are consumed in part by formation of neutrophil-platelet aggregates.37 Mucus Accumulation of excessive quantities of mucus in the large airways is characteristic of horses affected by RAO and can contribute to nonbronchospastic airway obstruction. Accumulation of mucus is attributable to decreased clearance and increased production. The mucus in horses with RAO differs in both composition and viscoelasticity from that of clinically normal horses and this might contribute to its decreased clearance. The viscosity of mucus can increase threefold in RAO susceptible horses stabled and exposed to hay dust. Increased production of mucus is associated with up-regulation of the equine MUC5AC gene, which is responsible for production of mucin, but not with IL-13 or epithelial gene (CLCA1, EGFR, Bcl-2 and MUC5AC expression,39 in the small airways of horses with RAO. Airway Function and Gas Exchange Inhalation of inciting agents causes changes in lung function characterized by an increase in pulmonary resistance, lower dynamic compliance, altered distribution of ventilation, impaired gas exchange, increased functional residual capacity, and an altered breathing strategy. Airway obstruction is a result of bronchospasm, inflammatory thickening of airways, and accumulation of mucus and cells in the airways. Bronchospasm is largely relieved by administration of bronchodilator drugs or removal of the inciting cause, but residual effects on lung function remain and are attributable to inflammation and fibrosis and bronchoconstriction of small airways. Bronchoconstriction in both normal and affected horses is caused by parasympathetic activity and release of acetylcholine that reacts with muscarinic receptors on airway smooth muscle. However, the response is exaggerated in horses with RAO. Stimulation of airway sensory receptors results in an exaggerated bronchoconstrictive response, possibly because of the action of inflammatory mediators and/or byproducts. The exaggerated bronchoconstrictive response is not specific for allergens, and any substance that activates airway sensory receptors may incite bronchoconstriction once the sensitivity of the receptors is enhanced by inhalation of the inciting allergens. Exaggerated airway responsiveness to inhaled irritants persists for up to 3 days after a single exposure to the inciting agent and is likely important in the development of clinical signs of the disease. Bronchoconstriction increases work of breathing, but hypoventilation probably contributes little to the hypoxemia of affected horses, given that Paco2 is rarely increased. Hypoxemia, which can be severe ( 6 weeks old.

Cough, slight nasal discharge. Rarely fever.

Eosinophils in tracheal aspirate.

Death rare.

Pneumocystis jirovici Immunodeficient foals (formerly P. or foals administered corticosteroids. carinii) pneumonia

Cough, mucopurulent nasal discharge, fever, lethargy, tachypnea.

Neutrophils and macrophages and P. jirovici cysts in tracheal aspirate or bronchoalveolar lavage fluid.

Pneumonia, diffuse Sulfonamide/trimethoprim with neutrophilic or 30 mg/kg q12 h lymphocytic/ recommended but plasmacytic often not effective. infiltration and alveolar edema. P. jirovici evident in silver-stained lung sections.

PCR, polymerase chain reaction.

treatment with antibiotics other than a macrolide (erythromycin, azithromycin, clarith­ romycin, gamithromycin) and rifampin is associated with a lower recovery rate. Treatment with penicillin, with or without gentamicin, chloramphenicol, or tetracycline, is not effective. Trimethoprim–sulfadiazine combinations might be effective in some foals but are not the preferred treatment. Neomycin has been recommended for treatment of R. equi pneumonia, but the risk of nephrotoxicosis, need for parenteral administration, and lack of demonstration of clinical efficacy do not support its use at this time. The treatment of R. equi pneumonia in foals is achieved by administration of macrolide antibiotics in combination with rifampin. Conventional treatment is administration of the combination of an acid-stable erythromycin (preferably estolate) at a dose of 25 mg/kg orally every 12 hours and rifampin at a dose of either 5 mg/kg every

12 hours or 10 mg/kg every 24 hours. Other esters or preparations of erythromycin are less well absorbed or have shorter elimination half-lives than the estolate ester and must be administered more frequently. Erythromycin ethylsuccinate does not provide optimal therapy for R. equi pneumonia in foals because of poor absorption after oral administration. The macrolide antibiotics azithromycin and clarithromycin have also been used to treat foals with R. equi pneumonia. Treatment of foals with a combination of clarithromycin (7.5 mg/kg orally every 12 hours) and rifampin results in improved survival over foals treated with azithromycin (10 mg/kg orally q24h) and rifampin or erythromycin and rifampin in a veterinary teaching hospital. Azithromycin is typically administered with rifampin at a dose rate of 10 mg/kg q24h for every 24 hours for 5 to 7 days and then once every 48 hours. Gamithromycin (6 mg/kg

intramuscularly [IM] or intravenously [IV] once every 7 days, with or without administration of rifampin) is currently not recommended for routine use pending results of studies demonstrating its equivalence to or superiority over other treatments. Administration of gamithromycin (6 mg/kg IM or IV once every 7 days, with foals administered IV gamithromycin also administered rifampin) was associated with resolution of lesions detected by ultrasonographic examination in 95% of foals with R. equi pneumonia.23 IM was associated with marked lameness in 35% of foals and colic that required administration of analgesics in 45% of foals. Tulathromycin was not as effective as the combination of azithromycin– rifampin in treatment of R. equi abscesses in foals in a large prospective field study.24 Tilmicosin is poorly active against R. equi.25 Ultrasonographic examination of the thorax of foals may permit identification of

1018

Chapter 12 

■ 

Abscess score (cm)

VetBooks.ir

TUL 7

Diseases of the Respiratory System

DOX

DOX-RIF

AZM-RIF

Placebo

P Treatment = 0.089 Time = < 0.001 Treatment × time = 0.348

6 5 4 3 2 1 0 0

1

2

3

6 4 5 Time (weeks)

7

8

9

10

Fig. 12-30  Abscess score based on ultrasonographic examination of foals on a farm with endemic R. equi infection administered a placebo, tulathromycin, doxycycline, doxycycline and rifampin, or azithromycin and rifampin. There was no effect of antimicrobial treatment on resolution of the abscess score. (Modified from Venner M, Astheimer K, et al.: Efficacy of Mass Antimicrobial Treatment of Foals with Subclinical Pulmonary Abscesses Associated with Rhodococcus equi, J Vet Int Med 27:171, 2013.)

foals with clinically inapparent pulmonary abscesses, providing the opportunity for early intervention in the disease. However, many (~90%) lesions less than 10 cm in diameter will resolve without treatment, and treatment with antimicrobials confers no clear benefit over watchful waiting (Fig. 12-30).12 Another study demonstrated resolution of lesions in 44% of foals with lesions greater than 1.0 cm administered a placebo—a resolution rate not statistically different from that in foals administered tulathromycin (IM), azithromycin alone, or azithromycin and rifampin.6 It is unclear if there is an advantage to combined therapy of rifampin and a macrolide compared with treatment with a macrolide alone. The current recommendation is to use the combination of drugs. Gallium maltoate has been investigated for treatment of foals with R. equi pneumonia. The pharmacokinetics in foals have been determined, and it appears to be safe for administration to foals and was not inferior to administration of clarithromycin and rifampin in treatment of foals with pulmonary lesions consistent with R. equi infection.26-30 However, the study did not include an untreated or placebo treated group, and given the high rate of spontaneous resolution of lesions without treatment in such foals,6,12,31 it cannot be concluded that either treatment was superior to no treatment. Therapy should be continued until the foal is clinically normal and has a normal plasma fibrinogen concentration and white blood cell count, which can require treatment for at least 1 month and often longer. Radiographic or ultrasonographic demonstration of resolution of the pulmonary consolidation and abscessation is useful in the decision to stop therapy. The casefatality rate is approximately 30% (see “Epidemiology”) even with appropriate treatment.

Adverse effects of macrolide–rifampin therapy include the development of diarrhea in some foals and their dams. Administration of erythromycin to foals is associated with an eightfold increase in the risk of diarrhea. Antibiotic therapy should be temporarily discontinued in foals that develop diarrhea. During hot weather, some foals treated with erythromycin become hyperthermic (40–41° C [104–105.5° F]) and tachypneic, and occasional deaths result from this syndrome. The basis for this hyperthermic event, which may occur in healthy foals administered erythromycin, is unknown. Affected foals should be treated urgently with antipyretics, cold water bathing, and housing in a cooler environment. The emergence of R. equi isolates resistant to rifampin and one or more macrolides has been documented and underscores the need for monitoring of R. equi sensitivity to these antimicrobials. Case-fatality rate is higher (75%) in foals with R. equi resistant to one or more of rifampin and a macrolide compared with that in foals infected with susceptible bacteria (30%).32 The development of resistance during monotherapy with rifampin is a recognized contraindication to the use of this drug alone. Ancillary therapy with NSAIDs, bronchodilators, and mucolytics might be of value. Foals in severe respiratory distress require intranasal or intratracheal administration of oxygen.

CONTROL

Control measures are designed to maximize the resistance of the foal to infection and to reduce the infection pressure on the foal by decreasing contamination of the foal’s environment with virulent R. equi. Ensuring adequate transfer of colostral immunoglobulins in all foals through routine monitoring of serum immunoglobulin concentrations in

1-day-old foals is an essential part of any control program. To decrease environmental contamination with virulent R. equi, efforts should be made to reduce fecal contamination of pastures and to reduce or eliminate dusty or sandy areas. These efforts should include grassing or paving of bare areas, removal and composting of fecal material on a regular basis, reduction of stocking density, and reduction in the size of mare/ foal bands. On farms with endemic disease, regular physical examination, including ultrasonographic examination of the thorax of foals and once-daily monitoring of rectal temperature, can permit early identification of affected foals. These foals can then be monitored for resolution or progression of the disease, with animals in the latter group administered antimicrobials. Comments noted earlier about the effectiveness of mass medication of all foals with lung lesions should be noted.6,12,31 Measurement of blood white cell count, as detailed previously, can be useful in early identification of affected foals. Identification of one foal affected with R. equi pneumonia on a farm should prompt an examination of all other foals on the farm. The administration to foals of a hyperimmune serum, obtained from mares vaccinated with an autogenous vaccine, limits the severity of disease produced by experimental challenge but has not been consistently useful in preventing or decreasing the prevalence of naturally occurring disease. This unpredictable efficacy could be attributable to variable concentrations of R. equi antiVap-A IgG in batches of plasma.33 There are no vaccines effective in prevention of R. equi pneumonia in foals.34,35 FURTHER READING

Vázquez-Boland JA, et al. Rhodococcus equi: the many facets of a pathogenic actinomycete. Vet Microbiol. 2013;167:9-33.

REFERENCES

1. Goodfellow M, et al. Equine Vet J. 2015;47:508. 2. Vazquez-Boland JA, et al. Vet Microbiol. 2013;167:9. 3. Rzewuska M, et al. Vet Microbiol. 2014;172:272. 4. Bolton T, et al. J Vet Diagn Invest. 2010;22:611. 5. Stoughton W, et al. J Vet Int Med. 2013;27:1555. 6. Venner M, et al. Vet J. 2012;192:293. 7. Cohen ND, et al. Am J Vet Res. 2013;74:102. 8. Cohen ND, et al. Am J Vet Res. 2008;69:385. 9. Cohen ND, et al. Am J Vet Res. 2012;73:1603. 10. Sanz M, et al. Vet Microbiol. 2013;167:623. 11. Sanz MG, et al. Vet Immunol Immunopathol. 2015;164:10. 12. Venner M, et al. J Vet Int Med. 2013;27:171. 13. Giguere S, et al. Vet Radiol Ultra. 2012;53:601. 14. Venner M, et al. Pferdeheilkunde. 2014;30:561. 15. Reuss SM, et al. JAVMA. 2009;235:855. 16. Johns IC, et al. J Vet Emerg Crit Care. 2011;21:273. 17. Reuss SM, et al. Vet Radiol Ultra. 2011;52:462. 18. Passamonti F, et al. Vet J. 2015;203:211. 19. Leclere M, et al. Vet J. 2011;187:109. 20. Witkowski L, et al. Vet Immunol Immunopathol. 2012;149:280. 21. Chicken C, et al. Equine Vet J. 2012;44:203. 22. Giguere S, et al. Vet Microbiol. 2015;178:275.

Diseases of the Equine Respiratory Tract

VetBooks.ir

23. Hildebrand F, et al. Pferdeheilkunde. 2015;31:165. 24. Venner M, et al. Vet J. 2007;174:418. 25. Womble A, et al. J Vet Pharmacol Ther. 2006;29:561. 26. Chaffin MK, et al. J Vet Pharmacol Ther. 2010;33:376. 27. Coleman M, et al. Vet Microbiol. 2010;146:175. 28. Martens RJ, et al. J Vet Pharmacol Ther. 2010;33: 208. 29. Chaffin MK, et al. Am J Vet Res. 2011;72:945. 30. Cohen ND, et al. J Vet Int Med. 2015;29:932. 31. Venner M, et al. Vet Rec. 2013;173:397. 32. Giguere S, et al. JAVMA. 2010;237:74. 33. Sanz MG, et al. Vet Rec. 2014;175. 34. Lohmann KL, et al. Can J Vet Res. 2013;77:161. 35. Giles C, et al. Equine Vet J. 2015;47:510.

STREPTOCOCCUS ZOOEPIDEMICUS INFECTION Streptococcus equi var. zooepidemicus (S. zooepidemicus) is one of the bacteria most commonly isolated from the upper respiratory tract of both clinically normal horses and horses with respiratory disease and from the female genital tract, wounds, and guttural pouch.1 Almost all horses harbor a number of antigenic types of S. zooepidemicus in their tonsils, and this may be the source of opportunistic infections of other body systems, including the lungs and genital tract. Currently, over 300 variants of S. zooepidemicus are recognized by multilocus sequence typing, and disease is associated with specific variants.2,3 S. zooepidemicus is the most common beta-hemolytic streptococcus isolated from horses at necropsy examination, representing 72% of isolates.4 Most isolates are from placenta, fetal tissues, and genital tract of mares, but this likely represents the population of animals examined and would not include clinically normal horses in which S. zooepidemicus is commensal in the upper respiratory tract. Outbreaks of upper or lower respiratory disease are associated with particular variants of S. zooepidemicus (eg, ST-24 and ST-307),2,5 and endometritis is caused by a specific variant genetically distinct to those causing respiratory disease.6 Pathogenicity of S. zooepidemicus in horses is related to the presence of superantigens (szeN and szeP, but not szeF).7 S. zooepidemicus can cause disease in humans, cats, dogs, and poultry.8-12 Infection and disease of humans working with horses by S. zooepidemicus identical to or closely related to that isolated from horses with which the human cases had contact highlights the zoonotic potential of the organism.12 An outbreak of disease in chickens was associated with a strain of S. zooepidemicus isolated from horses on the same farm,9 and infection of three dogs, with disease in two, housed on horse stud farms.8 The disease in dogs is usually a highly contagious often fatal pneumonia.11 The organism also causes acute, severe pneumonia and systemic illness in cats usually as an outbreak of disease in catteries.10

S. zooepidemicus is frequently isolated from horses with pleuropneumonia, endometritis, neonatal septicemia, abortion, and mastitis, suggesting a role for this organism in the pathogenesis of these diseases.4 S. zooepidemicus is likely important in the development of respiratory disease in foals and adult horses. S. zooepidemicus was isolated from 88% of foals with clinical evidence of lower respiratory tract disease, and isolation of the organism was associated with an increased proportion of neutrophils in bronchoalveolar lavage fluid, suggesting a causal role for this organism. Similarly, the number of S. zooepidemicus isolated from tracheal aspirates of adult horses is directly proportional to the number of neutrophils in the aspirate and the probability that they have a cough. The association of S. zooepidemicus and inflammatory airway disease in racehorses is independent of previous viral infection, suggesting a role for S. zooepidemicus as a primary pathogen. Presence and number of colony forming units (cfu) of S. zooepidemicus in tracheal aspirates of horses is significantly associated with the risk of the horse having inflammatory airway disease. Adult horses dying of pneumonia associated with transportation often yield S. zooepidemicus on culture of lung lesions, and the disease can be reproduced experimentally. S. zooepidemicus with Chlamydophila caviae causes conjunctivitis and rhinitis in adult horses.13 These results clearly demonstrate a role for S. zooepidemicus in the pathogenesis of respiratory disease of horses. However, it is unclear whether S. zooepidemicus is a primary cause of disease, a secondary contaminant, or an invader of airways compromised by viral infection or other agents. Clinical signs of S. zooepidemicus infection of the lower respiratory tract of foals and horses include coughing, mild fever, mucopurulent nasal discharge, and increased respiratory rate. Endoscopic examination of the trachea and bronchi reveals erythema and presence of mucopurulent exudate. Tracheal aspirates or bronchoalveolar lavage fluid of affected horses or foals have an increased (>10%) proportion of neutrophils. S. zooepidemicus is a frequent isolate from the cornea of horses with ulcerative keratitis. Treatment consists of the administration of antimicrobials, including penicillin (procaine penicillin, 20,000 IU/kg IM every 12 hours) or the combination of a sulfonamide and trimethoprim (15-30 mg/kg orally every 12 hours). S. zooepidemicus isolates from horses in southern England demonstrate increasing resistance to tetracycline but not the combination of trimethoprim and sulfonamide (TMS).14 Most S. zooepidemicus isolates (70%) are resistant to gentamicin, whereas 95% are sensitive to penicillin and 55% sensitive to TMS. Forty-five percent of isolates are resistant to enrofloxacin—a recent phenomenon.14 Different sensitivity patterns are reported for S. zooepidemicus

1019

isolates from Western Canada, although the high proportion of isolates sensitive to penicillin (95%) and ceftiofur (99%) is consistent with that in England.1 A higher proportion of Canadian isolates are sensitive to gentamicin (85%) or enrofloxacin (91%). Control consists of isolation to prevent spread of infectious respiratory disease and vaccination to prevent viral respiratory disease. FURTHER READING Waller AS. Equine respiratory disease: a causal role for Streptococcus zooepidemicus. Vet J. 2014;201:3-4.

REFERENCES

1. Clark C, et al. Can Vet J. 2008;49:153. 2. Lindahl SB, et al. Vet Microbiol. 2013;166:281. 3. Waller AS. Vet J. 2014;201:3. 4. Erol E, et al. J Vet Diagn Invest. 2012;24:142. 5. Velineni S, et al. Vet J. 2014;200:82. 6. Rasmussen CD, et al. Vet Res. 2013;44. 7. Rash NL, et al. Res Vet Sci. 2014;97:481. 8. Acke E, et al. Vet Rec. 2010;167:102. 9. Bisgaard M, et al. Avian Dis. 2012;56:561. 10. Blum S, et al. Vet Microbiol. 2010;144:236. 11. Priestnall S, et al. Vet J. 2011;188:142. 12. Pelkonen S, et al. Emerg Infect Dis. 2013;19:1041. 13. Gaede W, et al. Vet Microbiol. 2010;142:440. 14. Johns IC, et al. Vet Rec. 2015;176:334.

STRANGLES SYNOPSIS Etiology Streptococcus equi subsp. equi. Epidemiology Highly contagious disease that affects horses of all ages but is most common in young animals. Prolonged carrier state in asymptomatic animals. S. equi causes disease in only equids. Clinical signs Acute onset of fever, anorexia, depression, submandibular and pharyngeal lymphadenopathy with abscessation and rupture, and copious purulent nasal discharge. Metastatic infection in other organ systems. Clinical pathology Culture of S. equi from nasal and abscess discharges. Polymerase chain reaction (PCR) of nasal, pharyngeal or guttural pouch swabs. High serum antibody titer to SeM. Lesions Caseous lymphadenopathy with rhinitis and pharyngitis, pneumonia, and metastatic infection in severe cases. Diagnostic confirmation Culture of S. equi or PCR. Treatment Systemic administration of penicillin. Local treatment of abscesses. Control Isolation and quarantine of cases. Serologic testing followed by PCR and culture of nasopharyngeal swabs or guttural pouch lavage of serologically positive horses enabling detection of   carrier status. Vaccination might reduce   the case attack rate and severity of disease but confounds identification of carrier horses.

1020

Chapter 12 

■ 

Diseases of the Respiratory System

ETIOLOGY

VetBooks.ir

Streptococcus equi subsp. equi (S. equi) is a gram-positive coccobacillus that produces a beta-hemolysin, evident as a zone of clear hemolysis surrounding colonies growing on blood agar. There is evidence that S. equi is a biovar or genovar of S. zooepidemicus. S. equi is highly host-adapted to Equidae. Genetic analysis, particularly of the variable region of the SeM gene, demonstrates the existence of clones that vary geographically.1-5 For instance, 21 SeM alleles were detected among 145 S. equi strains isolated in the United Kingdom,1 and two SeM alleles detected in horses in New Zealand had distinct geographic distributions.5 Similar analysis reveals the presence of two major S. equi clades in Ireland, with both being also common in the United Kingdom.4 Individual outbreaks can be caused by S. equi of the same SeM type and be restricted geographically or by use of horse.6 Analysis of SeM mutations in real time allows differentiation or linkage of strangles outbreaks and enables risk assessment of equine events where there is incursion of the disease. It is unclear whether changes in the SeM protein associated with these strains are associated with differing pathogenicity.1 There is variation in virulence related to the amount of M protein and hyaluronic capsule produced. An atypical milder form of the disease is associated with a capsule-deficient variant of S. equi, and an intranasal vaccine is based on a live, attenuated, nonencapsulated SeM-2 strain, although this strain can cause disease.7

EPIDEMIOLOGY Occurrence Strangles occurs in horses, ponies, donkeys, and mules worldwide, with the exception of Iceland. Outbreaks are seen relatively frequently on breeding farms and in polo and racing stables, when the infection is introduced by new arrivals that are often asymptomatic, and in horses taken to fairs and riding schools. An incidence of 35% over a 3-year period is reported for horse studs in Australia, and there were approximately 600 recorded outbreaks in the United Kingdom in 2010.2 Strangles can affect horses of any age, although the morbidity rate is usually greater in younger horses such as foals and weanlings. Age-specific attack rates of strangles of 18% for brood mares, 48% for 1-yearold horses, and 38% for foals during an outbreak on a breeding farm are reported, although higher morbidity rates (100%) can occur, especially in young horses. The risk of occurrence of an outbreak of strangles increases with the size of the group of horses: farms with 100 or more horses have a 26 times greater risk of experiencing an outbreak than farms with fewer than 15 horses. The case-fatality rate without treatment is about 9%, but with adequate early

treatment it may be as low as 1% to 2%. Deaths are usually attributable to pneumonia. Source of Infection and Transmission S. equi is an obligate parasite of horses and all infections are attributable to transmission from infected horses, either directly or by fomites. Nasal and abscess discharge from infected animals that contaminates pasture, tack, stalls, feed and water troughs, grooming equipment, and hands and clothes of grooms and veterinarians is often the source of infection for susceptible horses. S. equi survives in the environment for less than 3 days, and although fomite transfer is important in transmission of infection, prolonged quarantine of facilities is not warranted.8 Direct transmission from infected animals to susceptible animals occurs through contact. Approximately 10% to 40% of horses that recover from the clinical disease have persistent infection of S. equi in the pharynx and guttural pouches for many months and are an important source of infection. Horses with clinically inapparent disease, such as some cases of guttural pouch empyema, can shed the organism for over 3 years. The period of infectivity is important in terms of the length of quarantine that needs to be imposed on horses that have apparently recovered from the disease. Because nasal shedding of S. equi can be intermittent, repeated culture of nasopharyngeal swabs or use of PCR examination of guttural pouch washings is necessary to document the carrier status of individual horses. The clinically inapparent nature of the infection makes detection of carriers problematic, especially when considering introduction of horses into a previously closed herd in which strangles is not endemic. Endoscopic or radiographic examination of clinically inapparent shedders can demonstrate lesions in the guttural pouches, paranasal sinuses, or pharynx, but because some persistent carriers of S. equi do not have detectable abnormalities of the nasopharynx, the most reliable approach to detecting carriers is PCR examination of nasal swabs or guttural pouch lavage fluid (see “Control”).9 Animal Risk Factors Strangles is more common in young or naive horses, although the disease can occur in horses of any age. Animals that have previously had the disease are less likely than naive animals to develop the disease on subsequent exposure. A proportion (approximately 25%) of horses that recover from the disease do not develop a protective immune response and are susceptible to reinfection and a second bout of strangles. Resistance to the disease is associated with the pro­ duction of serum and mucosal IgG antibodies to the streptococcal M protein. The presence in the nasopharynx of antibodies

to streptococcal M protein is thought to be important in conferring resistance to the disease. Serum IgGb antibodies specific for SeM protein, which is important in the antiphagocytic activities of S. equi, are produced by most but not all horses during convalescence. Similarly, IgA and IgGb against SeM protein are detectable on nasal and pharyngeal mucosa after S. equi infection but not after intramuscular administration of vaccines containing M protein. Serum bactericidal activity alone is not considered to be a good indicator of resistance to the disease, especially if it is induced by administration of a vaccine. Antibodies similar to those found in the nasopharynx after infection with S. equi are present in colostrum and milk of mares that have recovered from the disease, are passed to foals via the colostrum, and are secreted into the foal’s nasopharyngeal mucosa. These acquired antibodies are important in mediating the resistance of young foals to the disease. Although strong immunity occurs after an attack, this immunity wanes. Importance Strangles is one of the most important diseases of horses in developed countries, accounting for up to 30% of reported infectious disease episodes. The disease is important not only because of the deaths that it causes but more importantly because of the disruption of the management of commercial horse establishments, the time necessary to treat affected horses, and the esthetic unpleasantness of the running noses and draining abscesses.

PATHOGENESIS

Virulence of S. equi is attributable to the presence of M proteins on the surface of the bacteria, a hyaluronic acid capsule and the production of a leukocidal toxin. M proteins are associated with S. equi adhesion to oral, nasal, and pharyngeal tissues; invasion of pharyngeal tonsils and associated lymphoid structures; and evasion of the innate host immune response. S. equi produces two M proteins—SeM and SzPSe. SeM is unique to S. equi and plays a dominant role in resistance of the organism to phagocytosis. Variations in structure of M protein are associated with decreased virulence. The M proteins interfere with the deposition of complement component 3b on the surface of the bacteria and bind fibrinogen, both of which reduce the susceptibility of the bacteria to phagocytosis by neutrophils. The antiphagocytic activity of S. equi reduces the efficacy of neutrophils in engulfing and destroying the bacteria. The capsule of S. equi is associated with resistance to nonimmune phagocytosis and pathogenicity. Strains of S. equi that do not produce a capsule do not induce disease, although they are able to infect guttural

Diseases of the Equine Respiratory Tract

VetBooks.ir

pouches and cause seroconversion in experimental studies. Following exposure of the oral and nasopharyngeal mucosal surfaces to S. equi, bacteria lodge in the pharyngeal and tonsillar lymphoid tissues, where they multiply rapidly. There is no evidence of colonization of mucosal surfaces and streptococci can be detected in pharyngeal tonsils within hours of exposure.10 The binding of S. equi to pharyngeal cells is caused by fibrinogen binding proteins associated with M protein. The resistance of S. equi to nonimmune phagocytosis results in accumulation of large numbers of organisms surrounded by degenerating neutrophils. Release of streptolysin S and streptokinase may contribute to tissue damage by directly injuring cell membranes and indirectly through activation of plasminogen. Bacteremia may occur. Migration of neutrophils into the lymph nodes causes swelling and abscessation within 48 hours of infection,10 with associated disruption of lymph drainage and development of edema in tissues drained by the affected nodes. Swelling of retropharyngeal lymph nodes may interfere with deglutition and respiration. Most abscesses eventually rupture and drain, and the infection resolves with the development of an effective immune response. Nasal shedding of S. equi usually begins 4 to 7 days after infection, or 2 days after onset of fever, and persists for 2 to 3 weeks in most horses but up to years in exceptional cases. Cessation of shedding accompanies development of an effective serum and mucosal immune response. Death is usually attributable to pneumonia caused by aspiration of infected material, although other causes of death include asphyxiation secondary to upper airway swelling and impairment of organ function by metastatic infection. Rare deaths also occur as a result of infarctive purpura hemorrhagica in horses infected with S. equi. Metastatic infection of the heart valves, brain, eyes, joints, and tendon sheaths or other vital organs can occur and cause a chronic illness and eventual death. Metastatic infection may occur because of bacteremia or extension of infection along chains of lymph nodes. Purpura hemorrhagica can occur as a sequela to S. equi infection and is associated with high serum antibody titers to SeM.

CLINICAL FINDINGS

The disease manifests as an acute disease of varying severity, chronic infection of retropharyngeal lymph nodes and guttural pouches, and as chronic disease associated with metastatic infection of organs distant to the upper respiratory tract.11,12 The severity of the acute disease varies with the age and immune status of the animal, the size of the inoculum, and the duration of exposure to infection. The term strangles derives from the enlarged retropharyngeal lymph nodes and

guttural pouches causing respiratory distress in severely affected equids. Acute Disease The acute disease is characterized by mucopurulent nasal discharge and abscessation of submandibular and retropharyngeal lymph nodes. After an incubation period of 1 to 3 weeks the disease develops suddenly, with complete anorexia, depression, fever (39.5– 40.5° C [103–105° F]), a serous nasal discharge that rapidly becomes copious and purulent, and a severe pharyngitis and laryngitis. Rarely there is a mild conjunctivitis. Lymphadenopathy becomes apparent as the submandibular lymph nodes enlarge and palpation elicits a painful response. The pharyngitis may be so severe that the animal is unable to swallow, and there is a soft, moist cough. The head may be extended. The febrile reaction commonly subsides in 2 to 3 days but returns as the characteristic abscesses develop in the lymph nodes of the throat region. The affected nodes become hot, swollen, and painful. Swelling of the retropharyngeal lymph nodes can cause obstruction of the oro- and nasopharynx with subsequent respiratory distress and dysphagia. Death by asphyxiation can occur at this time in severe cases. Obvious swelling of the nodes can take 3 to 4 days to develop; the glands begin to exude serum through the overlying skin at about 10 days and rupture to discharge thick, cream-yellow pus soon afterward. Average cases run a course of 3 weeks; severe cases can last as long as 3 months. Retropharyngeal abscesses can rupture into the guttural pouches, resulting in guttural pouch empyema and ultimately in prolonged infection and formation of chondroids. Retropharyngeal lymph node abscessation might not be apparent on external evaluation and can often only be detected by radiographic or endoscopic examination of the pharynx. Infection of retropharyngeal lymph nodes and guttural pouches is important in persistent infection and carrier status of some horses. If the infection is particularly severe, many other lymph nodes, including the pharyngeal, submaxillary, parotid, and retrobulbar nodes, can abscess at the same time. Local abscesses also occur at any point on the body surface, particularly on the face and limbs, and the infection can spread to local lymphatic vessels causing obstructive edema. This occurs most frequently in the lower limbs, where edema may cause severe swelling. Abscess formation in other organs probably occurs at this time. An atypical form of the disease can occur and is characterized by widespread subclinical infection within a stable or yard and a mild disease. Affected horses have a transient fever for 24 to 48 hours and a profuse nasal discharge, and are anorexic. A moderate enlargement of the mandibular lymph nodes

1021

occurs in only about one-half of the affected horses. Strangles in burros is a slowly developing debilitating disease. At postmortem examination the characteristic lesions consist of caseation and calcification of abdominal lymph nodes. Complications Complications occur in about 20% of cases. The most common fatal complication is the development of suppurative necrotic bronchopneumonia, which probably occurs secondary to the aspiration of pus from ruptured abscesses in the upper airway, or metastatic infection of the lungs. Extension of the infection into the guttural pouches, usually as a result of rupture of retropharyngeal lymph nodes into the medial compartment, causes empyema, which can lead to the formation of accretions of inspissated pus (chondroids). Involvement of the guttural pouches is evident clinically as distension and, after resolution of other signs, unilateral or bilateral nasal discharge. Guttural pouches of affected horses should be examined endoscopically for evidence of retropharyngeal abscessation or guttural pouch empyema or chondroid formation. Retropharyngeal lymphadenopathy can impair the function of the recurrent laryngeal nerves, with subsequent unilateral or bilateral laryngeal paresis and consequent respiratory distress. Metastatic infection (“bastard strangles”) results in the formation of abscesses in any organ or body site but most commonly in the lungs, mesenteric lymph nodes,11,13 liver, spleen, kidneys, and brain. Clinical signs depend on the organ affected and the severity of the infection, but intermittent fever, chronic weight loss, and sudden death as a result of rupture of abscesses into a body cavity are common manifestations of metastatic infection. Rectal examination or percutaneous ultrasonographic examination can reveal intra-abdominal abscesses in some horses with metastatic abscesses in the abdomen. Peritoneal fluid from these horses is often abnormal. Metastatic infections can occur in the central nervous system. Extension of infection to the meninges results in suppurative meningitis characterized clinically by excitation, hyperesthesia, rigidity of the neck, and terminal paralysis. Abscesses in the brain cause a variety of clinical signs, depending on location of the abscess, including severe depression, head pressing, abnormal gait, circling, and seizures. Metastatic infections of the ocular and extraocular structures, heart valves and myocardium, joints, bones, tendon sheaths, and veins may occur. Purpura hemorrhagica can occur as a sequela to S. equi infection. Two myopathic syndromes occur with S. equi infection in horses. Muscle infarction,

1022

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

which may be extensive, is assumed to result from immune-mediated vasculitis associated with purpura hemorrhagica. Often the muscle lesions in these horses are associated with other lesions consistent with severe purpura hemorrhagica, including infarctions in the gastrointestinal tract, skin, and lungs. Rhabdomyolysis and subsequent muscle atrophy results in signs of muscle disease, including stilted gait and elevated serum activity of creatine kinase and other musclederived enzymes, and is assumed to be attributable to cross-reactivity of anti-SeM antibodies with myosin. Myocarditis and glomerulonephritis have been suggested as sequelae to S. equi infection but have not been conclusively demonstrated to occur.

CLINICAL PATHOLOGY

Hematologic abnormalities during the acute phase of the disease include leukocytosis, with a neutrophilia reaching a peak as the lymph nodes abscess. Hyperfibrinogenemia is characteristic of both the acute and chronic disease. Hematologic and biochemical abnormalities associated with metastatic infection depend on the site of the infection and its severity. Leukocytosis with a hyperproteinemia attributable to a polyclonal agammaglobulinemia is characteristic of metastatic and chronic abscessation. Hypoalbuminemia may be present. Serum biochemical profile can reveal evidence of specific organ dysfunction. There can be an anemia, which is likely attributable to the hemolytic effect of streptolysin O, immunemediated hemolysis, or anemia of chronic disease. A number of serologic tests to measure antibodies to SeM have been developed. An early commercial test that measured the serum IgG antibody titer to SeM was used to determine response to vaccination, suitability for vaccination and presence of metastatic infection. This ELISA has a sensitivity and specificity of 90% and 77%, respectively.14 The test is not useful in diagnosis of the acute disease. Serum antibody titers to SeM are very high (>1 : 12800) in horses with metastatic infection or purpura hemorrhagica. Further tests have been developed with the aim of detecting horses that have been exposed to S. equi, with the intent of enabling quarantine and control measures.14 ELISA assays for antibodies to SeM that combined analysis of two antigens restricted to S. equi provides sensitivity and specificity of 93% and 99%, respectively.14 Use of this assay allows detection of horses that have been exposed to S. equi and therefore might be carriers of the organism. These horses can then be examined using PCR of nasopharyngeal swabs (3 over 3 weeks) or guttural pouch lavage fluid (once).9,15,16 The high sensitivity of the test means that horses that test negative are unlikely to have been exposed or to be carriers.14

PCR testing is useful to detect shedding of S. equi DNA and has a greater sensitivity than routine culture.9,15,16 PCR testing of nasopharyngeal swabs or guttural pouch lavage fluid has a sensitivity of 90% to 95% and specificity of 86% to 97% with turnaround time of approximately 2 hours.15,16 The test is reported to be more specific than culture for detection of S. equi shedding. The PCR does not differentiate between live and dead S. equi, and false-negative results occur in the presence of large numbers of S. equi. Culture of nasal, pharyngeal, guttural pouch, or abscess discharge will usually yield S. equi in 30% to 40% of horses with active disease or in carriers.9 Abscesses can rapidly become contaminated with S. zooepidemicus, which can impede isolation of S. equi, although the two can be differentiated by culture or PCR analysis.17

NECROPSY FINDINGS

In the rare fatalities that occur, necropsy examination usually reveals suppuration in internal organs, especially the liver, spleen, lungs, pleura, and peritoneum. When the last is involved, it is usually as a result of extension from abscesses in the mesenteric lymph nodes. The microscopic changes of abscessation and suppurative lymphadenitis are uncomplicated. The widespread ecchymotic hemorrhages of purpura hemorrhagica are not specific to this infection, but S. equi should always be investigated as a potential cause of such lesions. Samples for Confirmation of Diagnosis • Bacteriology—swab of abscess wall, enlarged lymph node (CULT), or PCR

DIAGNOSTIC CONFIRMATION

Confirmation of strangles depends on the detection of S. equi from nasopharyngeal swabs, discharges from abscesses, or guttural pouch lavage by PCR or culture. As discussed previously, PCR has greater utility at detecting presence of the organism. Shedding of S. equi in nasal discharges begins 1 to 4 days after the onset of fever, and ruptured abscesses often become contaminated with Streptococcus zooepidemicus and S. equisimilis. History and clinical findings are usually highly suggestive of the disease, and classical cases of the strangles do not represent a diagnostic challenge. However, outbreaks of milder form of the disease are more challenging to diagnose, and confirmation is based on identification of the organism or demonstration of seroconversion. In acute disease, nasopharyngeal swabs or pus aspirates from abscesses can confirm S. equi infection. Because false negative culture results occur in 30% to 40% of cases, and qPCR has a sensitivity 94% and specificity

96%, and combining qPCR with culture will detect more than 90% of infected horses.9 Carriers are defined as horses shedding bacteria more than 6 weeks after clinical recovery. These horses will have serologic evidence of infection and can be detected by a series of at least 3 nasopharyngeal swabs at weekly intervals, or a single guttural pouch lavage ideally combined with a single nasopharyngeal swab, submitted for qPCR combined with culture. This will detect greater than 90% of carriers.18 Infection by Actinomyces denticolens caused submandibular abscessation in horses that can appear clinically similar to strangles. Diagnosis is based on bacterial culture.19,20

TREATMENT

The specific treatment of choice for S. equi infection of horses is penicillin, either as procaine penicillin G (22,000 IU/kg intramuscularly every 12 hours) or potassium or sodium penicillin G (22,000 IU/kg intravenously every 6 hours). Tetracycline (6.6 mg/kg intravenously every 12-24 hours) and sulfonamide–trimethoprim combinations (15-30 mg/kg orally or intravenously every 12 hours) can be efficacious but should only be used if penicillin cannot be administered. Aminoglycosides, such as gentamicin or amikacin, and the fluoroquinolones are not effective. Proportions of a small number (10) S. equi isolates from horses in southern England during 2007 to 2012 resistant to various antimicrobials were as follows: enrofloxacin 40%, gentamicin 80%, penicillin or ceftiofur 0%, trimethoprim–sulfonamide combination 20%, doxycycline 10%, oxytetracycline 0%, and resistant to three or more antimicrobials 20%.21 Similar sensitivities are reported for 22 isolates from Western Canada, with all isolates sensitive to ampicillin, ceftiofur, cephalothin, penicillin, erythromycin, amoxicillin-clavulanic acid, and no isolates sensitive to amikacin or neomycin. Approximately 80% of isolates were sensitive to TMS or tetracycline.22 Use of ceftiofur, a third-generation cephalosporin, in horses is discouraged on public health grounds.21

DIFFERENTIAL DIAGNOSIS See Table 12-16 for a list of differential diagnoses of infectious upper respiratory tract disease of horses. Pneumonia should be differentiated from pleuropneumonia associated with transport or other stress. Chronic weight loss as a result of metastatic infection should be differentiated from equine infectious anemia, parasitism, inadequate nutrition and neoplasia, especially gastric squamous-cell carcinoma, alimentary lymphosarcoma, and granulomatous enteritis.

Diseases of the Equine Respiratory Tract

1023

VetBooks.ir

Table 12-16  Differential diagnosis of diseases of the upper respiratory tract of horses Clinical signs Diagnosis and clinical pathology

Disease

Epidemiology

Respiratory tract

Other

Strangles (Streptococcus equi infection)

Incubation period 4–8 days. Course 10–21 days. Spreads by inhalation or ingestion. Mostly young horses in recently commingled groups. Long period (many months) of inapparent infection in some horses.

Copious, purulent nasal discharge. Cranial lymphadenitis and rupture. Moist cough. Obstruction of pharynx can cause dyspnea.

Severe illness with suppuration, fever. Atypical cases show involvement of other organs. Serious sequelae include pneumonia, metastatic spread of infection, mesenteric abscess or purpura hemorrhagica.

S. equi in nasal, pharyngeal or guttural pouch swabs, oropharyngeal pus. or lymph node abscess pus. PCR of nasal, pharyngeal or guttural pouch swabs. Serology to detect exposed horses. Leukocytosis. Hyperfibrinogenemia.

Equine viral arteritis (EVA)

Incubation period 1–6 days. Course 3–8 days. Some deaths.

Serous/purulent nasal discharge. Slight cranial lymphadenitis, cough. Conjunctivitis, purulent with edema or petechiae. Dyspnea.

Severe disease. Anasarca. Ventral edema, prepuce, legs, scrotum. May be diarrhea, jaundice. Up to 50% of mares abort.

Virus in blood at fever peak. Serology. Leukopenia.

Equine viral rhinopneumonitis (EHV-1)

Incubation period 2–10 days. Course 2–5 days. Cough may last as long as 3 weeks.

Serous/purulent nasal discharge. Slight cranial lymphadenitis, coughing, conjunctivitis. Mild respiratory disease; in young.

Abortion in mares. Virus may cause myelopathy.

Virus in nasal discharge or peripheral blood buffy coat. PCR of nasal discharge or blood. Tissue culture and serologic tests. Leukopenia. Virus in intranuclear hepatic inclusions of fetus.

Equine viral rhinopneumonitis (EHV-4)

Incubation period 2–10 days. Course 2–5 days. Cough may last as long as 3 weeks.

Serous/purulent discharge. Slight cranial lymphadenitis, coughing, conjunctivitis.

Mild respiratory disease; in young horses.

Virus in nasal discharge. Tissue culture and serologic tests. Leukopenia.

Equine influenza (H3N8 rarely H7N1)

Incubation period 2–3 days. Course 7 days. Cough may persist 3–4 weeks. Enzootic, worldwide (not Australia). Explosive outbreaks; 80%–100% morbidity in young.

Nasal discharge slight, serous only. Slight cranial lymphadenitis. Severe cough. No conjunctivitis and no respiratory distress.

Minimal extrarespiratory signs. Temperature 39–41° C (102–105° F).

Virus in nasal discharge. Good serologic tests available. Rapid ELISA test for viral antigen in nasal secretions. PCR of nasal secretions.

Equine rhinitis virus

Incubation period 3–8 days. Rapid spread, high morbidity (70%). Solid immunity after natural infection. Excreted in urine.

Pharyngitis, pharyngeal lymphadenitis, nasal discharge serous to mucopurulent. Cough persists 2–3 weeks.

Mild disease. Emphasis on coughing. Fever to 39.5° C (103° F).

Equine rhinitis virus on tissue culture. Serologic tests available.

Equine adenovirus

Many inapparent infections. High proportion of population serologically positive.

Mild respiratory signs in adults. Fatal pneumonia in Arabian foals with combined immunodeficiency.

Transient softness of feces. In mares can cause abortion without clinical illness.

Adenovirus in oropharyngeal swabs. Serologic tests available.

ELISA, enzyme-linked immunosorbent assay; PCR, polymerase chain reaction.

There is considerable debate about the treatment of horses with strangles. Folklore and anecdotal reports suggest that antibiotic treatment of horses with strangles is contraindicated because it promotes the development of metastatic infection. There is no experimental or empirical evidence to support this contention, and horses with strangles should be treated with therapeutic doses of an appropriate antibiotic, such as procaine penicillin, for a period of time sufficient to effect a cure, as appropriate. Treatment for S. equi infection depends on the stage of the disease, as follows:

• Horses with early clinical signs including fever, anorexia, depression, and purulent nasal discharge should be isolated and treated with therapeutic doses of penicillin for at least 5 days. The purpose of treatment is to prevent further development of the disease in the affected animal and to minimize environmental contamination with S. equi and transmission to other horses. Treatment should start as soon as clinical signs are observed, and the full course of treatment

should be completed to minimize the chances of recrudescence of the infection. Treatment at this stage causes rapid resolution of fever, anorexia, nasal discharge, and lymphadenopathy in individual horses and may abort an incipient outbreak of the disease in a stable or yard. However, treated horses may not develop a protective immune response and consequently may be at risk of reinfection if exposed to S. equi after completion of the course of treatment, leading one authority to

1024

VetBooks.ir









• •

Chapter 12 

■ 

Diseases of the Respiratory System

recommend that only severely affected animals be treated. Horses with submandibular lymph node abscessation but without other clinical abnormalities probably do not require antibiotic treatment. Such horses should be isolated and efforts made to aid maturation and rupture of affected lymph nodes. Systemic antibiotic therapy with penicillin is indicated in horses with advanced signs of strangles, including prolonged fever, depression, anorexia, or dyspnea resulting from retropharyngeal lymphadenopathy. Retropharyngeal abscessation frequently responds to antimicrobial therapy, although surgical drainage may be required in some instances. Horses with metastatic infection require systemic penicillin therapy in combination with specific therapy for the complication. Pulmonary and mesenteric abscesses are problematic because they are usually not amenable to surgical drainage, and prolonged antimicrobial therapy is required to attempt to effect a cure. Guttural pouch empyema requires either surgical drainage or repeated flushing of the affected pouch through the pharyngeal openings. Removal of pus and inspissated material in the guttural pouches can be achieved under endoscopic guidance. Alternatively, rigid or flexible indwelling catheters can be inserted for repeated flushing of the pouches with sterile isotonic electrolyte solutions (such as 0.9% NaCl) and topical medications. Substances and solutions that are irritating or injurious to mucus membranes, such as iodine,23 hydrogen peroxide, and similar irritant compounds, should not be infused into the guttural pouches. Combined topical and systemic administration of potassium benzyl penicillin may be beneficial. Chondroids can often be removed using wire snares. Horses with metastatic or guttural pouch infections are likely infectious and should be isolated. Treatment of purpura hemorrhagica is dealt with elsewhere. Management of horses that have been exposed to horses with strangles is controversial. Some authorities recommend treatment of such in-contact horses with penicillin until affected horses are isolated and no longer are a source of infection. However, close examination of exposed animals, including monitoring rectal

temperature, and treatment of horses at the first sign of illness is probably a more reasonable approach. Ancillary treatment consists of administration of nonsteroidal antiinflammatory drugs (NSAIDs) to reduce swelling and provide pain relief, application of hot poultices to encourage rupture of abscesses, provision of intravenous hydration in animals unable to drink, and wound care, including cleaning of ruptured abscesses and application of petroleum ointment to surrounding skin to prevent scalding. Horses with severe upper airway obstruction may require placement of a short-term tracheotomy.

CONTROL

All establishments that house multiple horses, and at which horses both enter and leave, should have biosecurity plans detailing the measures to be taken before new horses enter the facility. The principles of control measures include the prevention of transmission of S. equi from infected horses (cases or carriers) to susceptible animals and enhancement of resistance to infection and disease. There are two basic approaches to strangles prevention: eradication or control.24 The eradication approach aims to create and maintain a guaranteed disease-free state within the group and is most suited for closed herds. The control approach aims to reduce the frequency and severity of outbreaks but accepts that disease will occur from time to time. In many facilities with large numbers or frequent turnover of horses, such as large training yards or stud farms, a control approach may be more achievable than eradication.24 The approach to managing horses entering a facility in which S. equi infection is not present involves serologic testing of all horses before entry using an ELISA of known high sensitivity and specificity. Horses that are seropositive on arrival have prima facie evidence of exposure and are considered to be potential carriers of S. equi until demonstrated by PCR and culture to be negative for the organism. These horses are then screened for S equi carriage by guttural pouch lavage combined with a nasopharyngeal swab tested by qPCR and culture.9,15 Horses with negative or equivocal serology on arrival should not be admitted to the facility and should be retested 10 to 14 days later to establish serologic status and then either admitted to the facility if they are seronegative or screened for S equi carriage if they are seropositive.24 Any horses testing positive for carriage need to be treated (see previous discussion) and cured of infection before entry into the yard. Vaccination provides a useful adjunct to management changes, especially in groups of horses with open management systems, and may be more appropriate for yards aiming

for control rather than eradication.24 However, vaccination complicates interpretation of serologic screening of new arrivals because it is not possible at this time to differentiate between serologic responses to vaccination and infection. Prevention of Transmission Methods to control transmission of S. equi on affected premises are detailed in Table 12-17 and are as follows: • Infected animals should be isolated immediately. • All potential sources of fomites— including pails, brooms, grooming brushes, and blankets—should be thoroughly cleaned and disinfected and the bedding burned. Disinfection with phenolic compounds is preferred because they retain their activity in the presence of some organic matter, whereas bleach and quaternary ammonium compounds are inactivated by organic material. • Emergency prophylactic treatment, using injections of benzathine penicillin every 48 hours in foals and yearlings that are most susceptible, has been used but most treated animals develop strangles when the treatment is discontinued. This method of prophylaxis is not recommended. • People who care for affected horses should, ideally, avoid contact with susceptible animals. If this is not practical, then strict isolation protocols, including the wearing of protective boots and clothes that are changed between affected and normal horses, should be implemented. • Horses with elevated temperatures should have nasopharyngeal or guttural pouch swabs cultured. • As detailed previously, horses should be examined by nasopharyngeal swab or guttural pouch lavage to detect carriers. Carriers should be treated and demonstrated to be no longer carriers before being allowed access to potentially susceptible horses. Enhanced Resistance The majority of horses develop solid immunity to strangles after recovery from the spontaneous disease. This immunity lasts for up to 5 years in approximately three-quarters of recovered horses. Maximum resistance to disease probably requires both systemic and mucosal immunity to a variety of S. equi factors including, but not limited to, M protein. As noted previously, vaccination will result in positive results of serologic testing for exposure to S. equi. It is not possible at this time to differentiate between responses

Diseases of the Equine Respiratory Tract

VetBooks.ir

Table 12-17  Aims and associated measures used to control transmission of Streptococcus equi in affected premises and herds Aim

Measure

Prevent spread of S. equi infection to horses on other premises and to new arrivals on the affected premises

Stop all movement of horses on and off affected premises immediately and until the outbreak is controlled. Horses with strangles and their contacts should be maintained in well-demarcated quarantine areas. Clustering of cases in groups allow parts of the premises to be allocated as contaminated or clean.

Establish whether clinically recovered horses are carriers.

At least three nasopharyngeal swabs or washings taken at weekly intervals from all recovered cases and their contacts and examined by culture and PCR. Horses that are consistently negative are returned to the clean area.

Investigate apparently healthy horses from which S. equi is recovered.

Serology to determine exposure, with positive horses subject to nasopharyngeal swabbing (three times) or guttural pouch lavage and PCR and culture. Serologically negative horses should be retested in 10–14 days.

Eliminate S. equi from guttural pouches.

Treatment of guttural pouches, as detailed under “Treatment.”

Prevent infection of uninfected horses by S. equi from infected horses.

Personnel should have dedicated protective clothing when dealing with infected horses. Personnel should not deal with infected and uninfected horses. If this is not possible, then infected horses should be dealt with after uninfected horses. Strict hygiene should be implemented, including provision of disinfection facilities for personnel and diligent and thorough cleaning of stables and barns. If practicable, equipment should be destroyed after use with infected horses. Organic material should be removed from stables and then appropriate phenolic disinfectants or steam should be applied. This cleaning should be repeated. Feces and waste from infected animals should be composted in an isolated location. Uninfected horses should not be introduced to pastures used to house infected horses for 4 weeks. Water troughs should be disinfected daily. Horse vans should be thoroughly cleaned and disinfected after each use.

PCR, polymerase chain reaction. Source: modified from Sweeney CR et al. J Vet Intern Med 2005; 19:123–134.

to vaccination and responses to natural infection. This ambiguity confounds use of serologic tests in control of the disease. The benefits of potential increases in resistance to the disease induced by vaccination should be weighed against the restrictions this imposes on use of serologic testing in control programs. The efficacy of vaccination of adult horses with S. equi bacterins or M protein extracts of S. equi administered intramuscularly is controversial. Administration of M protein vaccines elicits an increase in the concentration of serum opsonizing antibodies but does not confer a high degree of resistance to natural exposure. However, in a controlled field trial, vaccination with an M protein commercial vaccine three times at 2-week intervals reduced the clinical attack rate by 50% in a population of young horses in which the disease was endemic. Horses vaccinated only once were not protected against strangles. A modified live vaccine induced a strong antibody response but caused substantial morbidity and some deaths among young ponies, highlighting the challenges with use of attenuated vaccines.25

Administration of a live, attenuated submucosal vaccine to mares appears to be safe.26 This result suggests that, in the face of an outbreak, vaccination might reduce the number of horses that develop strangles but will not prevent strangles in all vaccinated horses. A common vaccination protocol involves the administration of an M protein vaccine intramuscularly for an initial course of three injections at 2-week intervals, with further administration of the vaccine every 6 months in animals at increased risk of contracting the disease. On breeding farms, vaccination of mares during the last 4 to 6 weeks of gestation and of the foals at 2 to 3 months of age might reduce the incidence of the disease. The vaccines are administered by the intramuscular route and frequently cause swelling and pain at the injection site. Injection site reactions are usually less severe with the M protein vaccines. Injection into the cervical muscles may cause the horse to be unable to lower its head to eat or drink for several days—injection into the pectoral muscles is preferred for this reason. There are reports of purpura hemorrhagica, the

1025

onset of which was temporally associated with administration of a S. equi vaccine. Owners should be clearly warned of the limited efficacy and potential adverse effects of vaccination. The effect of vaccination in confounding interpretation of results of serologic testing used in control of the disease should be considered before horses are vaccinated. Foals that receive adequate high-quality colostrum from exposed or vaccinated mares have serum and nasopharyngeal mucosal immunoglobulins (IgGb) that provide them with resistance to S. equi infection. This passive immunity wanes at approximately 4 months of age. Vaccination of brood mares 1 month before foaling increases colostral IgG antibodies to M protein, and presumably serum and mucosal immunoglobulin concentrations in their foals, but the efficacy of this approach in preventing strangles in foals is not reported. An intranasal vaccine of an avirulent live strain of S. equi has recently been developed and appears useful. Use of the intranasal modified live vaccine can result in strangles caused by the vaccine strain.7 The vaccine is composed of a live variant (strain 707-27) that does not possess a capsule and is therefore avirulent when administered intranasally. Anecdotal reports suggest that recent manipulation of the genome by deletion of genes HasA and HasB, associated with formation of the capsule, has increased the genetic stability of the vaccine strain. The live attenuated vaccine should only be administered intranasally to healthy horses. The efficacy of the vaccine in field situations, safety in the face of an outbreak and in pregnant mares, incidence of adverse effects, and risk of reversion to virulence have not been reported. It should not be used in potentially exposed horses during an outbreak of the disease. Intramuscular injection of the vaccine results in the formation of abscesses. The vaccine should not be administered to horses concurrently with intramuscular administration of other vaccines because of the risk of contamination of needles and syringes with S. equi vaccinal strain and subsequent development of abscesses at injection sites. An experimental modified live vaccine administered intramuscularly to ponies conferred protection to experimental challenge.27 Vaccination by submucosal injection of a modified live vaccine is reported to provide short-lived (90-day) immunity to disease. The commercial form of the vaccine is administered into the submucosal tissues of the upper lip and is recommended for use in horses at moderate to high risk of developing strangles. At present there is no evidence of reversion of the vaccinal strain to virulence, and horses developing strangles subsequent to vaccination have all been infected with virulent strains of S. equi, apparently before

1026

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

development of immunity as a result of vaccination. The vaccine appears to be safe for use in pregnant mares.26 FURTHER READING

Mallicote M. Update on Streptococcus equi subsp equi infections. Vet Clin North Am Equine. 2015;31:27-35.

REFERENCES

1. Ivens PAS, et al. Equine Vet J. 2011;43:359. 2. Parkinson NJ, et al. Vet Rec. 2011;168. 3. Libardoni F, et al. Vet Microbiol. 2013;162:663. 4. Moloney E, et al. Irish Vet J. 2013;66. 5. Patty OA, et al. NZ Vet J. 2014;62:63. 6. Lindahl S, et al. Vet Microbiol. 2011;153:144. 7. Cursons R, et al. Vaccine. 2015;33:3440. 8. Weese JS, et al. Can Vet J. 2009;50:968. 9. Lindahl S, et al. J Vet Int Med. 2013;27:542. 10. Timoney JF, et al. Equine Vet J. 2008;40:637. 11. Whelchel DD, et al. Equine Vet Educ. 2009;21:131. 12. Whelchel DD, et al. Equine Vet Educ. 2009;21:135. 13. Mair TS, et al. Equine Vet J. 2011;43:123. 14. Robinson C, et al. Vet J. 2013;197:188. 15. Webb K, et al. Vet J. 2013;195:300. 16. North SE, et al. Equine Vet J. 2014;46:56. 17. Baverud V, et al. Vet Microbiol. 2007;124:219. 18. Waller AS. Vet Clin Equine. 2014;30:591. 19. Albini S, et al. Vet Rec. 2008;162:158. 20. Beck A, et al. Can Vet J. 2011;52:513. 21. Johns IC, et al. Vet Rec. 2015;176:334. 22. Clark C, et al. Can Vet J. 2008;49:153. 23. Sherlock CE, et al. Equine Vet Educ. 2007;19:515. 24. Slater J. Strangles—practical management of outbreaks. In: AVA/NZVA Pan Pacific Conference. Brisbane: Australian Veterinary Association; 2015:827. 25. Borst LB, et al. Am J Vet Res. 2011;72:1130. 26. Reinhold B, et al. Equine Vet Educ. 2010;22:40. 27. Robinson C, et al. Vaccine. 2015;33:1160.

GLANDERS SYNOPSIS Etiology Burkholderia mallei Epidemiology Contagious disease of solipeds (equids) and possibly camels. Important potential zoonosis. Clinical findings Acute or chronic form, and characterized by pneumonia and nodules or ulcers in the respiratory tract and on the skin. The disease is highly fatal. Clinical pathology Complement fixation test, mallein test, isolation of organism Necropsy findings Extensive bronchopneumonia in acute cases. Miliary nodules in internal organs and ulcerated nodules in skin and respiratory tract. Treatment and control Control is by slaughter of clinically affected and carrier animals detected by serologic or mallein tests. Rarely are affected animals treated, and if so it is by prolonged administration of antimicrobials.

ETIOLOGY

Burkholderia mallei, a gram-negative bacterium, is the causative organism of glanders.

It has close genetic and antigenic relatedness to Burkholderia pseudomallei. Isolates of B. mallei recovered from three continents over a period of 30 years have identical allelic profiles, but phylogenetic determination of strains can be achieved using molecular diagnostic techniques (for example, nextgeneration whole-genome sequencing and multiple-locus variable-number tandem repeats).1-3 Determination of phylogenetic relationships is a powerful tool for determining the source, and epidemiologic characteristics, of outbreaks of the disease. The only natural hosts of the organism are equids, with infection in other species being a result of transmission from infected equids. Humans in close contact with affected equids can be infected and develop an often fatal disease. Infection in humans is also caused through inadvertent exposure in laboratories. The organism is considered a category B biothreat (biologic warfare agent) by the Centers for Disease Control in the United States.4,5

EPIDEMIOLOGY Geographic Occurrence Glanders is restricted geographically to South America, eastern Europe, Asia Minor, Asia, and North Africa. Recent cases in Western Europe (Germany) are reported in horse imported from Brazil,6 where the disease is present,3 and in another horse, some years later, that was born in Germany.7 These cases highlight the need for vigilance in detection of glanders.6 Occurrence of outbreaks of the disease since 1986 is cataloged and available.8 The disease has reemerged, or at least been detected, recently in India and Pakistan.2,9,10 An outbreak in Bahrain was attributed to multiple introductions of infection, rather than simply one source.1 The disease was more widespread but has been eradicated from most countries. Glanders was an important disease when there were large concentrations of horses in cities and armies, but now has sporadic occurrence, or occurs in localized outbreaks, even in infected areas. Host Occurrence Horses, mules, and donkeys are the species usually affected. The disease can occur naturally in camels, although the number of reported cases is low, suggesting that camels are not particularly susceptible to infection.11 Humans are susceptible and the infection is often fatal. Carnivores, including lions can be infected by eating infected meat and infections have been observed in sheep and goats. Source of Infection and Transmission B. mallei is an obligate parasite and is readily destroyed by light, heat, and the usual disinfectants and is unlikely to survive in a

contaminated environment for more than 6 weeks. Infected animals or carriers that have made an apparent recovery from the disease are the important sources of infection. Carriers can be clinically normal and shed the organism for years. Chronic nodular lung lesions, which have ruptured into the bronchi, infect upper airway passages and nasal or oral secretions. Spread to other animals occurs mostly by ingestion, the infection spreading on fodder and utensils, particularly communal watering troughs, contaminated by nasal discharge or sputum. Rarely the cutaneous form appears to arise through contamination of skin abrasions by direct contact or from harness or grooming tools. Spread by inhalation can also occur, but this mode of infection is probably rare under natural conditions. Experimental Reproduction An experimental model for disease has been reproduced by intratracheal inoculation of horses with cultures of B. mallei. Horses showed fever within 24 to 48 hours of challenge followed by the progressive development of signs of respiratory distress with epistaxis and purulent nasal and ocular discharge. On postmortem there was lymphadenopathy, ulcerative lesions in the nasal septa, and pneumonia. Host and Pathogen Risk Factors Horses tend to develop the chronic form, mules and donkeys the acute form, but all types of equid and all ages are susceptible. The disease is more likely when animals are in a stressed state from heavy work, and animals that are poorly fed and kept in a poor environment are more susceptible. The stress associated with movement of a large number of horses can precipitate an outbreak with high mortality rates. In the few animals that recover, there is a long convalescence with the frequent development of the “carrier” state. Animals rarely make a complete recovery. Economic Importance The disease has little current economic importance, although the threat of horse movement reintroducing glanders into countries that have eradicated it is a concern. Zoonotic Implications Although humans are not highly susceptible, the infection can gain access through skin abrasions to produce granulomatous disease and pyemia. Infection can also occur from inhalation of infectious material. The case fatality is high. Horse handlers in general are at risk, and veterinarians conducting postmortem examinations without proper precautions are at particular risk. The organism is identified as a possible agent of bioterrorism.

Diseases of the Equine Respiratory Tract

PATHOGENESIS

VetBooks.ir

Invasion occurs mostly through the intestinal wall and a septicemia (acute form) or bacteremia (chronic form) is set up. Localization always occurs in the lungs but the skin and nasal mucosa are also common sites. Other viscera can become the site of the typical nodules. Terminal signs are in the main those of bronchopneumonia or, in acute disease, chronic wasting.

CLINICAL FINDINGS Acute Disease Acute disease presents with high fever, cough, and nasal discharge, with rapidly spreading ulcers appearing on the nasal mucosa and nodules appearing on the skin of the lower limbs or abdomen. Death as a result of septicemia occurs in a few days. Chronic Disease The disease is evident as fever, inappetence, weight loss, enlargement of submandibular lymph nodes, and exercise intolerance in almost all affected horses. Cough, dyspnea, and nasal discharge occur in approximately two-thirds of cases, and greater than 70% of cases have ulcers on the nasal septum or nodules and ulcers in the skin, usually of the legs.12 Three major manifestations are described, although one or more of all three can occur in the same animal: 1. Pulmonary 2. Skin 3. Nasal, although the chronic nasal and skin forms commonly occur together. Pulmonary Form of Disease The pulmonary form manifests as a chronic pneumonia with cough, frequent epistaxis, and labored respiration. Nasal Form of Disease In the nasal form, lesions appear on the lower parts of the turbinates and the cartilaginous nasal septum. They commence as nodules (1 cm in diameter), which ulcerate and may become confluent. In the early stages there is a serous nasal discharge that may be unilateral and that later becomes purulent and blood stained. Enlargement of the submaxillary lymph nodes is a common accompaniment. On healing, the ulcers are replaced by a characteristic stellate scar. Skin Form of Disease (“Farcy”) The skin form is characterized by the appearance of subcutaneous nodules (1-2 cm in diameter), which soon ulcerate and discharge pus of the color and consistency of dark honey. In some cases the lesions are more deeply situated and discharge through fistulous tracts. Thickened fibrous lymph vessels radiate from the lesions and connect one to the other. Lymph nodes draining the area become involved and may discharge

1027

to the exterior. The predilection site for cutaneous lesions is the medial aspect of the hock, but they can occur on any part of the body. Animals affected with the chronic form are usually ill for several months, frequently showing improvement but eventually either dying or making an apparent recovery to persist as occult cases.

older molecular diagnostic tests did not do so because of the close genetic relationship between these organisms, more modern tests do discriminate at a level that is clinically useful.13,19 Discussion of all the currently available tests is beyond the scope of this text, and readers are referred to recent publications.5,8

CLINICAL PATHOLOGY

Mallein Test The test is not generally recommended because of animal welfare concerns; however, it can be useful in remote endemic areas where sample transport or proper cooling of samples is not possible.17 The mallein test involves the intradermal injection of mallein, a purified or semipurified protein of B. mallei,20 into the subcutaneous tissues of the eyelid or lateral side of the neck. Mallein (0.1 mL of a 1.0 mg/mL concentration of mallein) is injected intradermally with a tuberculin syringe. Ideally, the thickness of the skin is measured using calipers before injection of mallein and 48 hours following injection. Some infected animals exhibit a general hypersensitivity reaction after inoculation. The mallein test can be negative in recently infected animals, in those with acute disease, and in advanced cases in horses.17 The mallein test has poorer sensitivity (~75%) than does serologic testing (Rose Bengal—90%, complement fixation—97%, and others).21

Chronic disease caused anemia and a moderate leukocytosis and neutrophilia.12 The principal tests used in the diagnosis of glanders are demonstration of presence of the organism by culture or detection of specific DNA (such as by PCR testing),13 the mallein test, or one of various serologic tests—complement fixation test,14,15 16 C-ELISA, immunoblot, Rose Bengal test, indirect hemagglutination, agar-gel immunodiffusion, indirect fluorescent antibody testing, counterimmune electrophoresis, and dot-ELISA.8 Details of test procedures are available in the OIE Manual on diagnostic tests and vaccines.17 All serologic tests are dependent on the host mounting an immune response to infection. Detectable immune responses might require a period of up to 2 weeks after infection to develop to the stage where they are detectable. The precise time depends on host factors and the characteristics of the particular serologic test. The intent of testing affects the test chosen for use. Tests intended to screen horses for international travel must have a high sensitivity, to avoid false negative results, but also high specificity, to ensure that there are few false-positive results. From the point of international movement of horses, tests should first have a high sensitivity to ensure that there are few false-negative results—with the potential for consequent transportation of infected animals—whereas detection of diseased animals in populations of horses in which the disease is rare demands tests with high specificity. The solution is often to first screen with tests of high sensitivity, such as the complement fixation test, followed by a test with much higher specificity (but often lower sensitivity), such as immunoblotting.16 The outcome of such serial testing is a high sensitivity and specificity. The diagnostic performance of various tests has improved with use of refined reagents (including use of recombinant or purified bacterial proteins or lipopolysaccharide,16 or antibodies) and optimized tests conditions,18 such as the temperature at which complement fixation tests are incubated.15 All serologic tests can be inaccurate for periods up to 6 weeks following performance of the mallein test. Molecular diagnostic techniques must discriminate between B. mallei and the closely related B. pseudomallei. Whereas

NECROPSY FINDINGS

In the acute form there are multiple petechial hemorrhages throughout the body and a severe catarrhal bronchopneumonia with enlargement of the bronchial lymph nodes. In the more common chronic form, the lesions in the lungs take the form of miliary nodules, similar to those of miliary tuberculosis, scattered throughout the lung tissue. Ulcers are present on the mucosa of the upper respiratory tract, especially the nasal mucosa and to a lesser extent that of the larynx, trachea, and bronchi. Nodules and ulcers may be present in the skin and subcutis of the limbs, which may be greatly enlarged. Local lymph nodes receiving drainage from affected parts usually contain foci of pus and the lymphatic vessels have similar lesions. Necrotic foci may also be present in other internal organs. B. mallei, and sometimes Arcanobacterium pyogenes, are isolated from infected tissues, and this is the main means of confirmation of diagnosis at necropsy.

DIAGNOSTIC CONFIRMATION

In live animals that could be carriers, the complement fixation test is used as the official test in most countries. The mallein test is used in those horses whose sera is anticomplementary.

1028

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

DIFFERENTIAL DIAGNOSIS • • • •

Epizootic lymphangitis Ulcerative lymphangitis Sporotrichosis Melioidosis • Strangles • Rhodococcus equi infection • Equine pleuropneumonia • Other causes of pneumonia

TREATMENT There is little information on treatment because control of the disease requires death of affected equids to prevent further spread of infection, and the granulomatous nature of the disease likely requires prolonged administration of antimicrobials capable of penetrating abscesses. Antimi­ crobial sensitivity of B. mallei isolates is reported.22 However, in instances in which highvalue animals are treated, a treatment protocol of enrofloxacin (8 mg/kg IV q24 h) and trimethoprim–sulfadiazine (32 mg/kg IV q24h) for 7 days, followed by enrofloxacin (4 mg/kg IV q24h) and trimethoprim–sulfadiazine (16 mg/kg IV q24h) for 2 weeks, and then 6 mg/kg doxycycline PO q12h for 9 weeks has been used. Treated horses responded within 1 week to treatment, with reduction of pyrexia and improved appetite. Nodules on the legs had resolved by week 3 of treatment. All 23 treated horses recovered and did not have evidence of disease recrudescence or a carrier status 1 year after the cessation of treatment.12

CONTROL

Control of glanders involves measures to reduce spread of the disease among equids in areas where the disease is endemic and eradication of the disease when desired or when the disease occurs as an emergency disease outbreak in areas where the disease is not endemic. Control of glanders is based on identification of infected animals by either serologic testing, intradermal mallein testing, or detection of the organism (culture or PCR) (see previous discussion). The mallein test and complement fixation test are the OIEapproved tests for glanders for the purposes of international movement of horses— noting the comments given previously about the characteristics of these tests. When attempting to identify infected animals, the delay in seroconversion or development of a positive mallein test after infection should be considered. Mallein testing can influence the sensitivity of subsequent serologic testing. If glanders is detected, or suspected, in area free of the disease, then the affected horse and contact animals should be promptly quarantined until their disease

status has been established. Eradication of the disease involves identification of infected animals with subsequent euthanasia and controlled disposal of these equids. Equids that could have been infected but that are negative on serologic or bacteriologic testing should have serologic tests repeated in 2 to 3 weeks. During this time, they should be quarantined. Complete quarantine of affected premises is necessary. A vigorous disinfection program for food and water troughs and premises generally should be instituted to prevent spread while eradication is being carried out. Carcasses of infected animals and contaminated or potentially contaminated bedding, feed, and tack that cannot be disinfected should be burned or deeply buried, consistent with local culture and laws. B. mallei is susceptible to most common disinfectants, including benzalkonium chloride, 1% sodium hypochlorite, 70% alcohol, and others.5 B. mallei does not persist in soil and is destroyed by exposure to sunlight or heating (>55° C >131 F for at least 10 minutes).5 B. mallei is a potential zoonosis that can cause severe illness and death in people. Barrier precautions, including the wearing of surgical masks, face shields, gloves, and gowns, are strongly recommended for people dealing with infected or suspect equids.5 There is currently no vaccine for glanders in animals or people.17 FURTHER READING Dvorak GD, Spickler AR. Zoonosis update—Glanders. JAVMA. 2008;233:570-577. Khan I, et al. Glanders in animals: a review on epidemiology, clinical presentation, diagnosis, and countermeasures. Transbound Emerg Dis. 2013;60:204-221.

REFERENCES

1. Scholz HC, et al. PloS Neglect Trop Dis. 2014;8. 2. Hornstra H, et al. Emerg Infect Dis. 2009;15:2036. 3. Silva KPC, et al. Pesquisa Veterinaria Brasileira. 2009;29:439. 4. Glanders, 2011. (Accessed 19.08.15, at .). 5. Dvorak GD, et al. JAVMA. 2008;233:570. 6. Elschner MC, et al. Equine Vet Educ. 2009;21:147. 7. Anon. Vet Rec. 2015;176. 8. Khan I, et al. Transbound Emerg Dis. 2013;60:204. 9. Malik P, et al. Ind J Anim Sci. 2009;79:1015. 10. Malik P, et al. Vet Ital. 2012;48:167. 11. Wernery U, et al. Emerg Infect Dis. 2011;17:1277. 12. Saqib M, et al. BMC Vet Res. 2012;8. 13. Janse I, et al. BMC Infect Dis. 2013;13. 14. Khan I, et al. Vet Rec. 2011;169:495. 15. Khan I, et al. Rev Sci Techn—OIE. 2014;33:869. 16. Elschner MC, et al. BMC Vet Res. 2011;7. 17. Glanders. OIE Manual of Diagnostic Tests and Vaccines, 2015. (Accessed 20.08.15, at .). 18. Sprague LD, et al. BMC Vet Res. 2009;5. 19. Schmoock G, et al. Acta Vet Scand. 2015;57. 20. de Carvalho MB, et al. BMC Vet Res. 2012;8. 21. Naureen A, et al. J Vet Diagn Invest. 2007;19:362. 22. Naureen A, et al. J Equine Vet Sci. 2010;30:134.

VIRAL INFECTIONS OF THE RESPIRATORY TRACT OF HORSES Viral respiratory tract disease is considered by veterinarians in the United States to be second only to colic among medical diseases in importance to the health and welfare of horses. The situation is likely similar in most developed countries and especially those in which equine influenza is endemic. Episodes of upper respiratory tract disease characterized by fever, nasal discharge, and cough are common in horses, especially young animals and horses housed in groups in stables and barns. An estimated 17% of equine operations in the United States have one or more horses affected by upper respiratory disease each year, and 1.5% of horses develop the disease every 3 months.1 Upper respiratory disease is most common in spring and least common in winter. Strangles was an uncommon cause of disease, occurring in only three horses per 1000 per 3 months. Viral respiratory disease is approximately three times more common in horses less than 5 years of age. With the exception of Streptococcus equi and possibly Mycoplasma spp., all the other known or suspected causes of nonparasitic infectious upper respiratory disease of horses are viral and include the following: equine herpesvirus types 1, 2, 3 (rarely), and 4; equine influenza virus; arguably, equine rhinitis virus types A-1 and B-1, 2, and 3; equine adenovirus; equine viral arteritis; and, historically, equine parainfluenza type 3 virus. Equine Hendra virus and African horse sickness cause signs of severe respiratory disease. There is minimal evidence that equine coronavirus causes respiratory disease in horses2,3 and there is evidence that the Middle Eastern respiratory syndrome (MERS) coronavirus, a disease of humans and camelids, does not cause disease in horses.4 Both s. equi and equine arteritis virus infection can be mild and lack outstanding clinical signs, thus closely resembling disease associated with some viral causes of upper respiratory tract disease. Therefore differentiation among diseases associated with these agents based on clinical signs and epidemiologic characteristics is difficult, and definitive diagnosis is only achieved through serologic or microbiological examination of blood or nasal discharge. Isolation and identification of a causative organism from nasopharyngeal swabs or airway washings of acutely affected horses provides a definitive diagnosis, although on occasion more than one potential pathogen may be isolated. Demonstration of seroconversion or a three- to fourfold increase in titer from serum samples collected during the acute and convalescent (usually 3 weeks after onset of clinical signs) phases of disease is persuasive evidence of infection. Immunofluorescence,

Diseases of the Equine Respiratory Tract

VetBooks.ir

enzyme-linked immunosorbent assay (ELISA) and polymerase chain reaction (PCR) tests may provide rapid diagnosis through detection of viral particles in nasal swabs and tissue specimens. The ability to determine the cause of an outbreak of upper respiratory disease in horses is enhanced by the use of multiple diagnostic tests and obtaining samples from more than one horse in an outbreak. However, definitive diagnosis of the cause of nasal discharge, cough, and fever is often not achieved. All the agents known to cause upper respiratory disease in horses are relatively sensitive to environmental influences, and spread of the agent is dependent on transmission from infected horses, either directly or on fomites. Introduction of an infected horse into a susceptible population of horses may result in an explosive outbreak of upper respiratory tract disease. Such events are common on stud farms and in racing stables, where relatively closed bands of horses are maintained for much of the year. The movement of horses over long distances may facilitate the introduction of pathogens to which the local population of horses is naive. The opposite situation occurs when young horses are introduced into larger bands of mixed aged animals, such as happens in racing stables or barns of pleasure horses. The younger, possibly naive, horse is then exposed to endemic pathogens to which the resident horses have developed resistance. Young horses are at particular risk of developing infectious disease of the upper respiratory tract. The diseases are usually a problem only in yearlings and 2-year-olds; young foals acquire a passive immunity from the dam and adults have acquired a permanent immunity through exposure or vaccination. In a horse population it is the average age and the mix of ages that largely determine its herd resistance, and when 30% to 40% of that population has not previously been exposed to infection then major outbreaks are likely. All of the diseases are transmitted by droplet infection, and over long distances, so that limitation of their spread is possible only by rigid isolation and intensive sanitary precautions, and even the best protected studs are likely to be infected from time to time. Parainfluenza-3 Virus Upper respiratory tract disease associated with equine parainfluenza-3 (PI-3) is characterized by a mild self-limiting disease that is not clinically distinguishable from the others in the group. The epidemiology and economic importance of disease associated with this agent is unknown.1 Equine Adenovirus Infection Two antigenic types of equine adenovirus, EAdV-1 and EAdV-2, are recognized that

have been associated with respiratory disease in foals and adult horses and diarrhea in foals, respectively.1 The virus causes fatal pneumonia in Arabian foals, and likely Fell pony foals, with severe combined immunodeficiency and has been isolated from otherwise apparently healthy foals with severe pneumonia, but its importance in clinical respiratory disease of immunocompetent foals is uncertain. EAdV-1 can be isolated or detected by PCR from healthy adult horses, although at a very low rate,5,6 and from 1% to 3% horses with signs of upper respiratory disease.7,8 Genomic analysis of EAdV-2 indicates markedly different lineage to that of EAdV-1.9 The virus is readily isolated from, or detected by PCR, in nasal swabs of approximately 50% of sick or healthy foals.6 Postparturient mares can shed the virus.6 Infection with EAdV-1 and EAdV-2 is worldwide. Serologic surveys differ in the proportion of seropositive horses, likely at least partially a result of the testing methodology, with serum neutralization tests yielding higher seropositive rates than ELISA tests.5 Approximately 80% of horses in New South Wales, Australia are positive by serum neutralization assay for either or both of EAvV-1 or EAdV-2.5 EAdV is considered to cause a mild respiratory disease with fever, coughing, nasal discharge, and conjunctivitis. Foals are assumed to acquire the infection from their dams, which secrete the environmentally stable virus in nasal discharge, urine, and feces. The virus is not associated with inflammatory airway disease in racehorses in England, but it has been associated with small outbreak of upper respiratory tract disease. Diagnosis can be made on cell smears taken from conjunctiva or nasal mucosa that reveal characteristic adenoviral intranuclear inclusion bodies. Serologic methods include serum neutralization, hemagglutination inhibition, complement fixation, ELISA, or precipitating antibody tests. The serum neutralization test is most accurate, but the hemagglutination inhibition test is most suitable for a screening test. Virus genetic material can be detected by specific PCR testing.5,7 No specific control measures are indicated for normal foals. Reovirus A reovirus, or a series of serotypes, cause mild upper respiratory tract disease of horses. Infection with these agents appears to be of little clinical or economic importance. REFERENCES 1. Radostits O, et al. Viral diseases characterized by respiratory signs. In: Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1307. 2. Pusterla N, et al. Vet Microbiol. 2013;162:228. 3. Oue Y, et al. J Vet Med Sci. 2013;75:1261. 4. Meyer B, et al. Emerg Infect Dis. 2015;21:181. 5. Giles C, et al. Vet Microbiol. 2010;143:401.

1029

6. Bell SA, et al. Equine Vet J. 2006;38:379. 7. Ataseven VS, et al. Res Vet Sci. 2012;92:324. 8. Pusterla N, et al. Vet Rec. 2013;172. 9. Giles C, et al. Vet Microbiol. 2015;179:184.

EQUINE INFLUENZA SYNOPSIS Etiology Influenza virus H3N8 (previously A/ equine 2) of two lineages (Eurasian and American) and numerous, evolving, strains. Currently circulating viruses are of the American lineage, Florida clades 1 and 2. H7N7 has not been identified as a cause of disease for decades. Epidemiology Short incubation period and highly contagious nature of the virus result in explosive outbreaks of disease. Viral shedding by subclinically affected horses is important for introduction of infection to populations. Prolonged carrier state is not recognized. Clinical signs Upper respiratory disease complicated by pneumonia. Abortion is not a feature of the disease. Clinical pathology None characteristic. Lesions Rhinitis, pneumonitis. Rarely causes death. Diagnostic confirmation Demonstration of virus in nasopharyngeal swab either by culture, enzyme-linked immunosorbent assay (ELISA), polymerase chain reaction (PCR), or membrane-bound immunoassay. Treatment Supportive care. There is no specific treatment. Control Quarantine to prevent introduction of the virus. Hygiene and disinfection to prevent fomite spread. Vaccination in enzootic areas with vaccine containing strains or antigens protective against currently circulating strains (Florida clade 1 and clade 2), to prevent clinical disease.

ETIOLOGY Equine influenza is associated with infection by influenza A virus—either equine influenza A/H7N7 or equine influenza A/H3N8 virus, members of the influenza virus A genus of the family Orthomyxoviridae. Influenza A viruses are typed according to the surface proteins—hemagglutinin (HA) and neuraminidase (NA) of which there are 18 HA subtypes (H1-H18) and 11 NA subtypes (N1-N11).1 Influenza virus is an RNA virus that has eight segments to its genome that encode 10 proteins. The hemagglutinin and neuraminidase proteins are used for antigenic characterization of virus strains. Mutations in these genes or poor-fidelity RNA copying results in changes in amino acid composition of viral proteins that can be detected by serologic tests (see “Clinical Pathology”) and that have important consequences for infectivity and pathogenicity of the virus.

1030

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Of the two serologically distinct subtypes of equine influenza virus, all reported outbreaks in the past three decades have been associated with strains of EIV-A/H3N8. There are no reports of disease associated with EIV-A/H7N7 in the past 35 years, and reports of seroconversion might be related to use of vaccines containing EIV-A/H7N7 antigen. There are no reports of other influenza viruses, such as the H1N1 avian virus, causing disease in horses, although the avian-like influenza A/I/Jilin89 (H3N8) caused severe disease and high mortality among horses in China in 1989 and there is a single report of avian H5N1 being isolated from sick horses, which also had serologic evidence of exposure to the virus, in Egypt during an outbreak of the disease in birds.2 Equine influenza H3N8 virus can infect dogs and cause serious disease and death.3,4 Canine influenza virus infection, which originated in horses, is now endemic in dogs populations in much of the world.3 Dogs are also susceptible to infection with equine influenza virus (Florida clade 1) when in close contact with horses infected with, and clinically ill from, the virus.5 Equine influenza virus H3N8 was isolated from one of ~400 healthy Bactrian camels sampled in Mongolia.6 The H3N8 virus can infect pigs but is not associated with disease;7 seals, in which it can cause a fatal respiratory disease; and birds.8,9 Experimental infection of cats with equine N3H8 influenza virus causes respiratory disease, and the infection can spread to in-contact cats.10 Canine influenza, N3N8, which is of equine origin, does not appear to pose a zoonotic risk.10 At this time, equine H3N8 virus does not appear to be an important zoonotic threat. Equine influenza H3N8 virus was first detected as a cause of respiratory disease in horses in 1963 in the United States. It subsequently became widely distributed, appearing in the United Kingdom in 1965, and evolved into multiple lineages and sublineages. There are two major lineages of EIVH3N8 that circulate in horse populations—a Eurasian lineage and an American lineage (the names of which do not reflect the current geographic distribution of the viruses). This divergence in the virus occurred in the early 1980s, and there has been subsequent evolution of the American lineage into Kentucky, Argentinian, and Florida sublineages, with the Florida sublineage composed of two clades—clade 1 and clade 2 (Fig. 12-31).11 For purposes of vaccine production, clade 1 is represented by A/eq/South Africa/04/2003-like or A/eq/ Ohio/2003-like viruses, and clade 2 is represented by A/eq/Richmond/1/2007-like viruses.12,13 The predominating virus lineage or strain varies from year to year and from region to region. Both of Florida clades currently cocirculate and coevolve worldwide. Viruses

of the Eurasian lineage have not been detected since 2005 (2015 OIE data).12,14 The important point is that there is continual change in the viral lineage or strain in some populations of horses and that constant monitoring of viral strains is vital for appropriate composition of vaccines and for molecular epidemiology. For instance, the majority of viruses from Europe (France, Italy and the United Kingdom) and North America characterized antigenically and/or genetically between January 2003 and April 2004 were of the American lineage. This continues to be the case with American lineage, Florida sublineage, clade 2 being the virus detected in Europe,15 Northern Africa,16 Asia (India, China, Mongolia),17-19 Ireland (before 2009), and the United Kingdom.12,16 Within clade 2, there are at least two identified virus subpopulations with amino acid substitutions in HA1 at either position 144 or position 179. The majority of 2014 viruses characterized had a valine at position 144 and an isoleucine at position 179.13 Equine influenza viruses detected in recent outbreaks in the United States of America have been Florida sublineage clade 1,12 although clade 2 was isolated from a horse in California that had been imported from Europe.20 Although clade 1 viruses predominate in America and clade 2 in Europe, clade 1 viruses have caused outbreaks in Europe (France),15,21 Australia, Africa, and Asia, and the virus circulating in Ireland in 2014/15 is from clade 1 (although different from the virus that caused outbreaks in Australia and Japan in 2007).22 Viral Evolution Identification of the lineage and sublineage of the virus is based on nucleotide sequencing of the hemagglutinin gene to detect mutations in the gene resulting in amino acid substitutions in the HA1 domain. These amino acid substitutions alter the charge, acquisition of glycosylation sites, and/or receptor binding avidity of the virus and hence its biologic activity including infectivity, immunogenicity, and virulence.11 Hemagglutination inhibition assay (HI) has been used to type viruses, but this is now being complemented by genetic testing and determination of amino acid composition of major antigens (HA and NA). For instance, the amino acid composition of clade 1 and clade 2 viruses differs by at least seven amino acids in the HA1 domain of hemagglutinin.23 Information about EIV strains changes constantly and is available at the equiflunet or OIE websites.12,13 The existence of lineages and strains of virus is important in the epidemiology of the disease because the antigenic differences among strains can be sufficient to prevent cross-protection provided by natural infection or vaccination. Cross-protection refers to the ability of one antigen (virus strain) to produce immunity in the horse against

infection with another type of antigen (virus strain). Infection or challenge with the same type of antigen is referred to as homologous challenge, whereas that with a different antigenic type is referred to as heterologous challenge. Strains of influenza virus circulate between and among populations of horses, with more than one strain of virus circulating at any one time in some horse populations, although individual disease outbreaks are associated with a single viral strain. Many, but not all, of these virus strains are constantly evolving, and evolution of the viruses is necessary for perpetuation of cycles of infection through the emergence, or reemergence by cycling, of heterologous strains. Evolutionary stasis, the continued circulation of older strains of virus, occurs and has importance for vaccine composition for many diseases, but not, apparently, for equine influenza virus (EIV), where emergence of new strains is common and of great importance for control of the disease. Evolution of strains of equine H3N8 virus occurs through antigenic drift. Antigenic drift, the accumulation of point mutations in the gene coding for the major surface protein hemagglutinin, occurs continuously in virus circulating in horse populations. Antigenic drift occurs most rapidly in hemagglutinin protein but also occurs in M and NS genes. Antigenic drift, by producing heterologous viral strains, contributes to the continuing susceptibility of horses to infection and the reduced efficacy of some vaccines.11 For example, the 2007 outbreak of equine influenza in Japan in a population of vaccinated horses, was associated with the Florida clade 1 virus, whereas the vaccines in use at that time included viruses of Eurasian and American (Argentinian) strains.24 Antigenic shift is an event in which there is a dramatic alteration in the viral genome occurring by reassortment of viral genes during coinfection of a cell by two different types of virus (for example infection of a pig by both avian and human influenza viruses). Antigenic shift, which has not been documented for influenza viruses infecting horses, has the potential to produce new viruses with markedly different host infectivity and pathogenicity to either parent virus. RNA viruses, such as equine influenza virus, are genetically labile, and during an outbreak there is considerable genetic variation of the viruses infecting a single animal, with dominant and one or more less dominant variants of a strain proliferating in the horse and being transmitted to other horses.25,26 Furthermore, the dominant form of the virus within a horse can change over the course of the infection. This pattern of multiple variants infecting one horse and being transmitted to other horses results in a relatively large number of variants of the virus in a group of horses during an outbreak, constituting a loose bottleneck to viral evolution.25,26

Diseases of the Equine Respiratory Tract

1031

VetBooks.ir

Fig. 12-31  Phylogenetic tree illustrating evolving nature of equine influenza virus N3H8 from the virus originally detected in 1963. Note that there are two lineages (Eurasian and American) that diverged in the early 1980s; that the American lineage further evolved into Kentucky, Argentinian, and Florida sublineages (clades 1 and 2); and that these continue to evolve. Strains causing substantial outbreaks are highlighted in green, dates of large outbreaks are identified in the boxes, and vaccine strains are identified in yellow. (Reproduced with permission.11)

1032

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Persistence in the Environment According to the Ausvet Plan, equine influenza virus is inactivated by exposure to ultraviolet light for 30 minutes, by heating at 50° C (122 F) for 30 minutes, and by ether and acid (pH 3) treatment.27 Exposure to sunlight for 15 minutes at 15° C (59 F) also inactivates the virus. The virus persists in canal water (pH 6.9) for up to 18 days at 22° C (72 F) and 14 days at 37° C (98.6 F); in tap water (pH 7.0) for 14 days at 4° C (42 F) and up to 2 days at 37° C (98.6 F); in horse blood for 18 hours at 37° C (98.6 F); in horse urine (pH 8.0) for 5 to 6 days at 4° C (42 F), 15° C (59 F), and 37° C (98.6 F); in soil under dark storage at 18° C (65 F) for 24 hours; and in soil exposed to sunlight for 8 hours at 15° C (59 F).27 The capacity for the virus to persist in carcasses is unknown.27

EPIDEMIOLOGY Occurrence Worldwide, the only large horse populations in which influenza virus infection does not occur are in Australia and New Zealand, although Australia experienced its first outbreak of equine influenza in 2007 and subsequently was declared free of the virus.28,29 Widespread use of aircraft to move horses between countries in short periods has increased the spread of equine influenza viruses, as exemplified by the 2007 outbreak in Australia allegedly associated with importation of horses and failure to contain the infection in the quarantine facility;30 the 2003 outbreak in South Africa associated with a virus from North America;, and an earlier outbreak in Hong Kong. In all cases virus was introduced by imported horses. Epidemics of equine influenza have occurred in Europe or North America in 1956 (H7N7), 1963 (H3N8), 1969, 1979, and 1989, although this does not represent a comprehensive listing of large-scale outbreaks. Epidemics affecting more than 1 million horses occurred in China in 1989 (associated with the novel H3N8 Jilin virus) and 1993/1994 (associated with a conventional H3N8 virus closely related to 1991 European isolates). Epidemics in Japan, Europe, and North America have been associated with introduction of a novel virus (for example, the 1963 appearance of H3N8 virus in Miami) or antigenic drift of existing viruses and resultant inefficacy of extant vaccines.14,31,32 The epidemics in Australia (2007) and South Africa (2003) were associated with introduction of the virus into a naïve and unvaccinated population of horses.30 Localized outbreaks of disease in stables or race courses occur almost annually in countries in which the disease is endemic, likely related to the movement of horses into the training and racing populations, with subsequent introduction of virus and development of disease in at risk horses (see “Animal Risk Factors”). Disease associated with equine influenza virus usually occurs as

outbreaks associated with the introduction of virus into a population of susceptible horses. Virus may be introduced by clinically affected horses or, more commonly, by horses that are not noted to be clinically ill. Vaccinated horses can become infected and shed influenza virus while not becoming ill,33 especially if vaccinated with heterologous strains, and this is likely a common method of introduction of virus into susceptible populations. Outbreaks of influenza virus infection can cause clinical disease in nearly all (98%) horses in a susceptible population, although in populations of horses of mixed age and with varying serum titers to equine influenza the morbidity rate can be much lower (16%28%). The incidence of disease in one race track population was approximately 130 cases per 1000 horses at risk per month, although this rate likely varies widely among outbreaks. The mortality rate is usually very low (50%) in untreated and treated horses. There are no estimates of cause-specific mortality or morbidity rates.

PATHOGENESIS

The molecular pathogenesis of EMPF is unknown, although parallels are drawn between Epstein-Barr virus associated interstitial (fibrosing) pneumonia in people and the disease in horses.6 The lesion in horses progresses through proliferation of type 2 pneumocytes to alveolar fibrosis and focal obliteration of normal lung architecture. There is systemic evidence of inflammation, and the fever, weight loss, lethargy, and exercise intolerance of affected horses demonstrate a systemic response to the disease. Exercise tolerance could be attributable to diminished gas exchange in damaged lungs, the systemic inflammatory effects of the disease, or, more likely, a combination of both.

CLINICAL SIGNS

Horses with EMPF have various combinations of weight loss, recurrent cough, depression, anorexia, fever, tachycardia, tachypnea, or respiratory distress.5,7,8,22,27 Signs of respiratory disease might not be apparent at

Diseases of the Equine Respiratory Tract

VetBooks.ir

initial examination, but as the disease progresses respiratory distress develops in most, but not all, cases. The usual history is of a gradual onset of increased respiratory effort, although some horses have a sudden onset of respiratory distress. Heart and respiratory rates are often elevated. Pyrexia is not a constant finding and can be intermittent in affected horses. There can be a nasal discharge but this is not invariable or characteristic. Thoracic auscultation might reveal only increased intensity of normal breath sounds or the presence of occasional crackles and wheezes. Typically, there is tachypnea with an increased respiratory effort. Thoracic radiography reveals pulmonary disease, usually apparent as severe, diffuse interstitial disease with nodular opacities.7 The interstitial opacity can be diffuse or nodular with multiple well-defined opacities against an overall background of increased interstitial density. Ultrasonographic examination often reveals the presence of multiple nodules in the lung parenchyma confluent with the pleural surface.7 There is no excess pleural fluid. Lymphoma in horses with EMPF and lymphoma associated with EHV-5 infection and treated with acyclovir have been reported, raising the possibility of a common etiology of the two diseases.19,20

CLINICAL PATHOLOGY

Hematologic examination usually reveals a neutrophilic leukocytosis, mild anemia, lymphopenia, and hyperfibrinogenemia.2,7,8,22 Pancytopenia occurs in a small proportion of cases.2 Hypoproteinemia and hypoalbuminemia are common. Arterial oxygen tension is not invariably abnormal but declines as the disease progresses. Examination of a tracheal aspirate reveals neutrophil inflammation. Macrophages contain occasional intranuclear inclusions.7,8,28 Serologic testing for antibodies to fungi including Blastomyces, Coccidioides, Histoplasma, Aspergillus, and Cryptococcus spp. assists with ruling out diseases caused by these organisms. EHV-5 can be detected by PCR examination of bronchoalveolar lavage fluid in most affected horses.7

NECROPSY FINDINGS

Gross lesions are restricted to the lungs and occur in two distinct forms, the more common form being numerous coalescing nodules of fibrosis with little unaffected lung present (diffuse nodular form).1 Individual nodules are up to 5 cm in diameter, pale tan to white, and moderately firm. The less common lesion consists of multiple discrete nodules up to 10 cm in diameter and separated by grossly normal lung (discrete nodular form). The nodules are otherwise similar in appearance and texture to those of the diffuse form. Bronchial lymph nodes may be markedly enlarged.

Histopathologic findings are restricted to the lungs and bronchial lymph nodes, and the lesions are similar regardless of the gross pathology. Nodules are sharply demarcated from unaffected lung tissue, and they consist of marked interstitial expansion of alveolar parenchyma by well-organized mature collagen.1,7 In most cases, the alveolar architecture is preserved, but in rare cases, fibrosis is arranged in broad interlacing bundles without preserving the alveolar structure. Affected alveoli are lined by cuboidal cells, and the lumen contains inflammatory cells, primarily neutrophils and macrophages, the latter occasionally containing intranuclear inclusion bodies consistent with a herpesvirus infection. Changes in bronchial lymph nodes consist of marked lymphoid hyperplasia, often with nonspecific sinus histiocytosis. Specimen for Laboratory Diagnosis Specimens for diagnosis include lung nodules for histopathology, in situ hybridization, and PCR. DIFFERENTIAL DIAGNOSIS The differential diagnoses include the following: lung abscess, chronic pleuropneumonia, silicosis, lipid pneumonia, eosinophilic pneumonia, fungal pneumonia, pulmonary neoplasia (either primary—granular cell tumor—or secondary such as metastatic squamous-cell carcinoma), congestive heart failure, or chronic kidney disease.

DIAGNOSTIC CONFIRMATION Diagnosis is confirmed by demonstration of compatible lesions in lungs at necropsy or on biopsy.

TREATMENT

There are no treatments with established efficacy, and management of the disease is based on first principles and empirical treatment with antiviral drugs, antiinflammatory drugs, and antimicrobials. Reduction of inflammation and relief of pain is achieved by administration of nonsteroidal antiinflammatory drugs (phenylbutazone, flunixime meglumine, ketoprofen) or corticosteroids (dexamethasone or prednisolone). Antimicrobials are administered to treat secondary bacterial infection and include penicillin, penicillin in combination with an aminoglycoside, or tetracycline or doxycycline. Antiviral drugs have been administered to horses with EMPF, and some of these treated horses have survived.7,28 Acyclovir and valacyclovir (a metabolite of acyclovir) are both active in vitro against gammaherpesviruses. Acyclovir is administered orally (20 mg/kg PO q8h) but has variable absorption compared with valacyclovir, and one cannot be confident that adequate

1043

concentrations in the blood are achieved in all horses.29-31 The preferred drug, based on pharmacokinetic properties, is valacyclovir (30-40 mg/kg PO q8h).30,31 A 2-week course of treatment with valacyclovir was associated with resolution of the disease in one horse.28

CONTROL

There are no known control measures. FURTHER READING Dunkel B. Pulmonary fibrosis and gammaherpesvirus infection in horses. Equine Vet Educ. 2012;24:200.

REFERENCES

1. Williams KJ, et al. Vet Pathol. 2007;44:849. 2. Hart KA, et al. Equine Vet Educ. 2008;20:470. 3. Marenzoni M, et al. J Vet Diagn Invest. 2011;23:802. 4. Back H, et al. Acta Vet Scand. 2012;54. 5. Spelta CW, et al. Aust Vet J. 2013;91:274. 6. Williams KJ, et al. PLoS ONE. 2013;8:e77754. 7. Wong D, et al. JAVMA. 2008;232:898. 8. Schwarz B, et al. Acta Vet Hung. 2013;61:319. 9. Kubiski SV, et al. JAVMA. 2009;235:381. 10. Williams KJ. Vet Pathol. 2014;51:372. 11. Fortier G, et al. Vet J. 2010;186:148. 12. Diallo IS, et al. Arch Virol. 2008;153:1643. 13. Fortier G, et al. Vet J. 2009;182:346. 14. Ataseven VS, et al. Transbound Emerg Dis. 2010;57:271. 15. McBrearty KA, et al. NZ Vet J. 2013;61:254. 16. Hue ES, et al. J Virol Meth. 2014;198:18. 17. De Witte FG, et al. J Vet Int Med. 2012;26:1064. 18. Herder V, et al. Vet Microbiol. 2012;155:420. 19. Schwarz B, et al. Equine Vet Educ. 2012;24:187. 20. Vander Werf K, et al. J Vet Int Med. 2013;27:387. 21. Bawa B, et al. J Equine Vet Sci. 2014;34:694. 22. Niedermaier G, et al. Vet Rec. 2010;166:426. 23. Soare T, et al. Vet Rec. 2011;169:313A. 24. Dunowska M, et al. NZ Vet J. 2014;62:226. 25. Panziera W, et al. Brazilian J Vet Pathol. 2014;7:17. 26. Marenzoni ML, et al. J Vet Diagn Invest. 2011;23:802. 27. Soare T, et al. Vet Rec. 2011;169:313. 28. Schwarz B, et al. Equine Vet Educ. 2013;25:389. 29. Garre B, et al. Antimic Agents Chemother. 2007;51:4308. 30. Maxwell LK, et al. J Vet Pharmacol Ther. 2008;31:312. 31. Garre B, et al. J Vet Pharmacol Ther. 2009;32:207.

EQUINE HENDRA VIRUS INFECTION ETIOLOGY

An acute disease of horses transmissible to humans and characterized in horses by fever and respiratory distress, but with capacity for pleiotropic clinical expression, occurs in northeastern Australia. The disease is associated with infection by equine Hendra virus (henipavirus, HeV, in the family Paramyxoviridae), which is closely related to Nipah virus (classified as the same genus).1,2 There is very little genomic variation in HeV.3 Infection by HeV, or Nipah virus, causes meningoencephalitis or, less frequently, respiratory disease in humans in contact with infected horses.4 A disease syndrome in horses and humans in the Philippines in 2014 was

1044

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

associated with infection by a henipavirus closely related to Nipah virus.5 The disease caused encephalitic signs and death in both horses and humans, there was horse–human and human–human spread, and the source of infection appeared to be fruit bats. The case-fatality rate in humans was approximately 50%, with a higher fatality rate among those with an acute encephalitic disease. At least 10 horses died, although this number likely underrepresents the actual number of horse deaths. Infection in some humans was associated with butchering or eating horses.5

EPIDEMIOLOGY

The disease in horses is uncommon, in that the morbidity or mortality rate within the population of at risk horses is low, with approximately two to four outbreaks reported each year involving a small number of horses. Between 1994 and 2013 there were 48 outbreaks of disease in horses, of which approximately 6 involved human disease.2 The case-fatality rate was high in early outbreaks, and because of control measures that involve test and slaughter of infected horses, all horses infected in outbreaks are destroyed. The disease is important because of the zoonotic nature of the infection and the high case-fatality rate in infected humans. Transmission The source of the virus is a wildlife host, the frugiverous pteropoid bats (fruitbats and flying foxes, Pteropus spp.). Approximately 25% of pteropid bats, including representatives of all four main species in eastern Australia (the grey-headed flying-fox, Pteropus poliocephalus; the black flying-fox, Pteropus alecto; the little red flying-fox, Pteropus scapulatus; and the spectacled flying-fox, Pteropus conspicillatus), were identified as being seropositive for HeV. The bats are seropositive for antibodies to the virus, the only seropositive mammals of 34 wildlife species sampled, and the virus can be isolated from pteropoid postpartum uterine fluid and fetal tissue. Mechanism of spread from bats to horses is uncertain, but it is speculated that ingestion by horses of infected bat fetal fluids and tissues might transfer infection from bats to horses. Fruit bats are consistently present when the disease occurs in horses.6 The disease spreads from bats to horses, and there is considerable interest in determining risk factors associated with transmission.7-11 Infection of horses likely involves contact with virus soon after (hours) it is excreted from bats.9 This is consistent with the 40× increase in risk of disease for horses in postal codes where fruit bats roost.8 Serologic evidence indicates waxing and waning infection on a seasonal basis, and epidemiologic evidence and modeling favors an effect of anthropogenic changes in bat habitat favoring urbanization of bat colonies and

reduced migration of bats. Urbanization increases the risk of spread of infection to horses, and reduced migration of bats reduces herd immunity in flocks, resulting in outbreaks of virus shedding and spread to horses.12,13 Dissemination of infection between horses by mechanical spread of infected nasal discharge likely occurred in the largest outbreak, and this could have been the route of infection of the human fatality. The virus is present in nasal discharges and urine of infected horses, and spread from horse to horse might also occur through inhalation of infected urine. Horse-to-horse transmission of infection is uncommon,6 likely because the virus does not persist in the environment but can occur. Human-to-human transmission of infection has not been reported. Disease occurs in horses, humans, cats, and guinea pigs, although in the latter two species the disease was a result of experimental infection. Dogs can become infected, but they do not appear to be at high risk of developing the disease, if they are at any risk at all, and there is no evidence that they propagate infection. Fruit bats do not develop clinical disease when experimentally infected. Zoonotic Potential The disease has important zoonotic implications; there have been four human deaths (~60% case-fatality rate) as a result of meningoencephalitis or pneumonitis and respiratory failure. Deaths all occurred in people who had close contact with infected horses, and the high risk associated with treating infected horses, or performing postmortem examinations on horses that have died of the disease, has prompted some veterinarians in endemic areas to exit from equine practice.14 The reasons are concern about personal safety or legal liability for the safety of coworkers and owners of horses. However, the virus is not easily transmitted to humans, as evidenced by the observation that most people in contact with clinically affected horses do not develop antibodies to the virus.

CLINICAL SIGNS

The incubation period of the spontaneous disease is 8 to 11 days, but it is much shorter in experimentally induced disease. Death usually occurs within 24 to 48 hours of first onset of clinical signs, and affected horses housed in paddocks are often found dead.6 Clinical signs of the disease in horses include lethargy, which is often marked, depression, loss of appetite, fever, ataxia, blindness, head pressing, aimless wandering, tachycardia, tachypnea, and copious frothy nasal discharge. Horses can show aimless pacing and can become entangled in fence— which can be mistaken for an accident rather than a consequence of neurologic disease associated with HeV infection.6 There can

also be hemorrhagic nasal discharge and swelling of the head. Some horses have muscle tremor. Death in acutely affected horses is sometimes associated with severe respiratory distress. Clinically inapparent infections of horses can occur. An important understanding is that HeV can cause protean clinical signs, which might be interpreted as evidence of respiratory (dyspnea), neurologic (ataxia, blindness), muscular (muscle fasciculations), hepatic (head pressing) or gastrointestinal (terminal colic) disease.

CLINICAL PATHOLOGY

Characteristic changes in the hemogram or serum biochemical profile are not reported. If infected animals survive more than a few days after the onset of clinical signs, they develop serum-neutralizing antibodies. The recommended range of samples for HeV exclusion from the live horse are 10 mL of clotted, EDTA, and heparin blood; pooled nasal swabs from each nostril; swabs from other mucosal surfaces (e.g., oral cavity, rectum, or conjunctiva); or urine collected in, preferably, phosphate-buffered glycerol saline or isotonic sterile saline.6 Antibodies are detectable by immunofluorescence microsphere immunoassays, or rapid immune plaque assay.15 Viral genome can be detected by RT-PCR that is highly specific. Viral isolation in Vero cells or imaging using electron microscopy demonstrate presence of the virus. Details of diagnostic tests are available from the OIE.

NECROPSY

Necropsy examination reveals pulmonary edema with hemorrhage and froth in the airways. Histologic examination reveals an interstitial pneumonia characterized by extensive vascular damage and necrosis of alveolar macrophages. Pulmonary vascular changes include edema and hemorrhage within alveoli, plus necrosis and thrombosis of alveolar capillaries and small arterioles. The distinctive histologic feature is the presence of syncytial giant cells within blood vessels of the lungs and other organs. Retrospective diagnosis of the disease can be documented using an immunohistochemical technique or demonstration of viral nucleic acid in tissue by a test based on the PCR. Postmortem, 10  mL of blood can be collected from the jugular vein in addition to the submandibular lymph node and swabs as per a live horse. Field experience suggests that it is relatively easy to safely collect jugular blood from recently dead horses.6

TREATMENT AND CONTROL

There is no specific treatment for this disease. Ribavirin has been investigated for use in infected or exposed humans but is not used in horses, for which control measures are implemented.2

Diseases of the Equine Respiratory Tract

VetBooks.ir

The control measures in the described outbreaks included slaughter of all infected horses, extensive serologic testing, and control of movement of horses within a defined disease control zone. The disease in index cases is likely attributable to contact of susceptible horses with infected fluids of pteropoid bats, and interventions that prevent or reduce the frequency of this occurrence are sensible, although the efficacy of this control technique has not been determined. An effective vaccine is available, and its use is strongly advised in horses living or visiting areas where the disease is endemic.16-18 In addition to preventing disease in horses, the vaccine provides veterinarians attending horses in endemic areas with some level of confidence that the horse is not infected with HeV.17 Strict biosecurity measures must be used by veterinarians examining potentially infected horses in areas where the disease is endemic, although this practice is often met with resistance.18 Because of the protean nature of the disease, all sick horses should be considered as sources of infection. Biosecurity practices should be in place for examination of all horses, and the degree to which personal protective equipment is used can be adjusted based on the risk that the horse being examined is infected. Detailed guidelines for personal biosecurity are available.19 REFERENCES

1. Croser EL, et al. Vet Microbiol. 2013;167:151. 2. Aljofan M. Virus Res. 2013;177:119. 3. Marsh GA, et al. Emerg Infec Dis. 2010;16:1767. 4. Nakka P, et al. Clin Radiol. 2012;67:420. 5. Ching PKG, et al. Emerg Infect Dis [Internet]. 2015;21. 6. Ball MC, et al. Aust Vet J. 2014;92:213. 7. Smith C, et al. PLoS ONE. 2014;9. 8. McFarlane R, et al. PLoS ONE. 2011;6. 9. Martin G, et al. J Gen Virol. 2015;96:1229. 10. Field H, et al. PLoS ONE. 2011;6. 11. Edson D, et al. PLoS ONE. 2015;10. 12. Plowright RK, et al. Proc Royal Soc B. 2011;278:3703. 13. Breed AC, et al. PLoS ONE. 2011;6. 14. Mendez DH, et al. Emerg Infec Dis. 2012;18:83. 15. McNabb L, et al. J Virol Meth. 2014;200:22. 16. Pallister JA, et al. Virology J. 2013;10. 17. Middleton D, et al. Emerg Infec Dis. 2014;20:372. 18. Mendez DH, et al. BMC Vet Res. 2014;10. 19. Australian Veterinary Association guidelines for veterinary personal biosecurity, 2013. (Accessed 14.09.15, at .).

PULMONARY AND SYSTEMIC ASPERGILLOSIS (ASPERGILLUS SPP.) Diseases of horses and cattle associated with infection with Aspergillus spp. are characterized by either localized infections with slow progression or fulminant systemic or pulmo-

nary disease. Localized infections are of the nasal cavities and paranasal sinuses;1-3 eye; reproductive tract, including placenta;4 mediastinum; or guttural pouch (see “Guttural Pouch Mycosis”). Systemic disease can affect any organ, including the brain, liver, and kidney,5 but the most common manifestation is as acute pulmonary disease with or without infection of other tissues.3,5-9

ETIOLOGY

The causative organism is Aspergillus spp., usually A. fumigatus but occasionally one of A. flavus, A. deflectus, A. nidulans, A. niger, A. clavulatus, A. nidulans, or A. sydowii.3,5,7,8,10,11 Aspergilli reproduce both sexually and asexually and hence are classified as dimorphic fungi. Asexual reproduction is by production of conidiophores and conidia. The organism is ubiquitous in organic material, and infections are opportunistic and associated with heavy contamination with the organism or decreased host defenses, although obvious risk factors are not always identified. Because its ubiquitous, the organism is often recovered from tracheal aspirates performed using contaminated equipment in horses with mild signs suggestive of noninfectious respiratory disease, such as heaves. In this instance recovery of the organism is of no clinical importance.

EPIDEMIOLOGY

Risk factors for development of aspergillosis include heavy environmental contamination with conidia and decreased host resistance, such as in horses with immune suppression associated with myeloproliferative disease (lymphoma), enterocolitis, or administration of immunosuppressive drugs such as corticosteroids. Specific risk factors for guttural pouch mycosis and infections of the nasal cavity or paranasal sinuses have not been identified, with the exception of an association between surgical resection of ethmoidal hematoma and subsequent nasal aspergillosis. Systemic or pulmonary aspergillosis is commonly associated with rumenitis, thirdcompartment ulceration in camelids,7 enterocolitis, or administration of immunosuppressive drugs in adult horses. An outbreak pulmonary aspergillosis causing death of five albino Asinara donkey foals aged 20 to 30 days, but not of nonalbino herdmates, occurred without history of intercurrent disease or drug administration.6

CLINICAL FINDINGS

Aspergillus spp. causes both localized and systemic disease in horses, cattle, camelids, and likely other species. Localized diseases include guttural pouch mycosis, which is discussed in detail elsewhere in this text. Fungal granulomas in the paranasal sinuses or nasal passages in any species of farm animal are caused by a number of organisms, including Cryptococcus neo­ formans, Conidiobolus spp., Rhizomucor

1045

pusillus, Scedosporium apiospermum, and, rarely, Aspergillus spp.1-3,11-13 The disease is evident as nasal discharge that is usually unilateral, distortion of the contour of the head over the affected sinus, and lesions detectable on endoscopic examination of the nasal passages. Radiography can reveal the presence of a mass in the paranasal sinuses or nasal cavity associated with lysis and proliferation of bone. There is hyperfibrinogenemia and leukocytosis. Systemic aspergillosis, including aspergillus pneumonia, is a severe disease usually evident as acute death without localizing signs in animals with other preexisting systemic disease, such as enterocolitis, neonates with inadequate passive immunity, or those receiving immunosuppressive drugs.3,8,9 Horses with aspergillus pneumonia often have a very brief clinical course once signs of respiratory disease develop. Most commonly, horses with pulmonary aspergillosis die without signs of respiratory disease. Signs of pulmonary aspergillosis include fever, tachypnea, crackles and wheezes on thoracic auscultation, epistaxis, and frothy nasal discharge. Radiography reveals diffuse, miliary, nodule interstitial pneumonia (Fig. 12-32). Ultrasonographic examination demonstrates numerous small intrapulmonary masses adjacent to the pleural surface. Affected horses have hyperfibrinogenemia and leukocytosis at the time of development of the disease, but usually they have had neutropenia as a result of the enterocolitis. Aspergillus spp. can be isolated from tracheal aspirates of affected horses. The prognosis is very poor. Aspergillus fumigatus can cause solitary, cavitated lesions in the lungs of foals.8 Disseminated aspergillosis has a variety of manifestations but is always a severe disease with a brief clinical course. Affected horses often have severe depression and can have signs of brain disease as a result of mycotic vasculitis and encephalitis.5 The prognosis is very poor. Aspergillus spp. is also associated with development of granulomas in the mediastinum of horses without apparent predisposing factors. Affected horses have progressively worsening respiratory distress, cough, fever, and occasional nasal discharge. Horner’s syndrome can develop if the mass encroaches on the vagosympathetic trunk within the thorax. The mass is evident on radiographic examination of the thorax. Cultures of tracheal aspirates yields Aspergillus spp. Affected horses have neutrophilia, hyperfibrinogenemia, hyperglobulinemia, and mild anemia. Keratomycosis attributable to Aspergillus spp. infection is infrequent in horses. The disease is characterized by blepharospasm, photophobia, epiphora, and corneal ulceration and opacity. Aspergillus spp. infections of the reproductive tract include mycotic placentitis and abortion and mycotic

1046

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Fig. 12-33  Granulomatous lesion caused by R. seeberi in a Belgian Warmblood horse. (Reproduced with permission.1)

There are no specific control measures or means of preventing disease associated with Aspergillus spp. REFERENCES

Fig. 12-32  Radiograph of the caudal thorax of an adult horse with pulmonary aspergillosis secondary to acute enterocolitis. Note the military and interstitial densities.

endometritis.4 Fungal osteomyelitis of the proximal sesamoid occurs in horses that have received intraarticular administration of corticosteroids.14

CLINICAL PATHOLOGY

Definitive diagnosis of the disease is based on demonstration of organisms within lesions, either by histologic examination, culture, or use of PCR to demonstrate fungal DNA.2 Antemortem demonstration of high concentrations of antibodies to Aspergillus spp. provides persuasive, but not definitive, evidence of infection. Both agar gel immunodiffusion assays and ELISA assays are available. These assays might not be useful in immunocompromised animals or in those with fulminant disease.

NECROPSY FINDINGS

Acute lesions are characterized by purulent, necrotizing inflammation. Chronic lesions are granulomas that contain macrophages, neutrophils, and giant cells. Pulmonary lesions are characterized by an acute necrohemorrhagic alveolitis. Organisms morphologically consistent with Aspergillus spp. are detected in the lesions as fungal hyphae, although these must be differentiated from Pseudoallescheria boydii or Fusarium spp. Reagents for immunofluorescent detection of Aspergillus spp. in lesions are available and useful in confirming the diagnosis.

TREATMENT AND CONTROL

Treatment of systemic or pulmonary disease is usually unrewarding, although surgical

resection of a single large cavitating lesion in the lungs of a foal followed by administration of voriconazole (10 mg/kg PO q24h for 2–4 weeks) effected a cure.8 A dose of voriconazole of 4 mg/kg orally q24h produces concentrations of drug greater than 0.5 µg/mL in body fluids. This concentration is greater than the concentration of voriconazole required to inhibit growth of filamentous fungi.15 Localized disease can be treated by surgical resection and administration of antifungal agents. Antifungal agents reported to be effective in treatment of localized disease in horses associated with Aspergillus spp. include itraconazole (3 mg/kg q12h, PO for 3–5 months) or enilconazole (0.2%–2.0% solution administered topically via an indwelling intranasal catheter q12h for 2–5 weeks). The lesions were debulked before treatment with enilconazole was started.1 Seven of eight horses with nasal aspergillosis treated in this way recovered.1 Topical treatment with enilconazole (10 mg/mL of solution) after surgical resection resulted in resolution of aspergillosis of the frontal sinus of a horse. Topical administration of nata­ mycin (25 mg) was used for varying periods of time to treat mycotic rhinitis in three horses. Amphotericin is likely effective against Aspergillus spp. and is cheaper than the azole class of drugs, but it is potentially nephrotoxic and must be administered intravenously. Fluconazole is not effective against the filamentous fungi, including Aspergillus spp.

1. Kendall A, et al. J Vet Int Med. 2008;22:1239. 2. do Carmo PMS, et al. J Comp Pathol. 2014;150:4. 3. Breuer W, et al. Schw Arch Tierh. 2015;157:407. 4. Moretti A, et al. Large Anim Rev. 2013;19:155. 5. Headley SA, et al. Mycopathologia. 2014;177:129. 6. Stefanetti V, et al. J Equine Vet Sci. 2015;35:76. 7. Hughes K, et al. J Vet Diagn Invest. 2008;20:672. 8. Hilton H, et al. J Vet Int Med. 2009;23:375. 9. Breshears MA, et al. Vet Pathol. 2007;44:215. 10. Lee SK, et al. J Equine Vet Sci. 2012;32:835. 11. Fiske-Jackson AR, et al. Equine Vet Educ. 2012;24:126. 12. Ubiali DG, et al. J Comp Pathol. 2013;149:137. 13. Tremaine WH, et al. Equine Vet J. 2001;33:274. 14. Sherman KM, et al. JAVMA. 2006;229:1607. 15. Passler NH, et al. J Vet Pharmacol Ther. 2010;33:35.

RHINOSPORIDOSIS Rhinosporidiosis is a chronic disease of the nasal mucosa in cattle and nasal mucosa, pharynx, and larynx in horses that causes formation of large polyps or granulomatous lesions (Fig. 12-33).1,2 The causative agent, Rhinosporidium seeberi, is an aquatic protist that typically causes disease in amphibians. Its exact taxonomy is the subject of debate.2,3 Exposure to the organism is almost universal, based on serologic studies, in buffalo, cats, cattle, dogs, goats, horses, and in some areas with high prevalence of the disease, humans (such as Sri Lanka).4,5 The disease is not endemic in western Canada or the United Kingdom, and affected horses in those areas were imported from Argentina.2,6-8 The disease is reported in a Warmblood horse in Belgian that had never left the country.1 Other cases in horses are reported from the Costa Rica, the southern United States, South Africa, and South America.7,9 The disease is evident as single or multiple, pedunculated or sessile, pink to red masses in the mucous membranes of the

Diseases of the Swine Respiratory Tract

VetBooks.ir

nose and nasopharynx. The lesions can bleed and become evident as epistaxis.7 No uniformly effective treatment is described, and surgical removal in three horses with pharyngeal or laryngeal lesions was not associated with cure—the disease progressing slowly over many months.1,2,7 Excision of a single mass in the rostral nares of a mule was curative.9 There is no effective pharmacotherapy. Confirmation of disease is achieved by examination of biopsy material demonstrating moderate multifocal hyperplasia and ulceration of the mucosa, mild to moderate, multifocal, lymphoplasmacellular inflammatory infiltrate multiple and spherical to polygonal organisms of variable appearance, consistent with R. seeberi, in the lamina propria mucosae.7 PCR analysis of affected tissue reveals presence of R. seeberi DNA.2,7 A related condition in cattle also thought to be caused by an unidentified fungus, similar to Rhinosporidium spp., is nasal granuloma, in which the lesions are small (0.5– 2.0 cm diameter) mucosal nodules in the anterior third of the nasal cavity. Histologically, there is a marked eosinophilic reaction, and yeast-like bodies are present in cells or free in the tissue spaces. Clinical signs include severe dyspnea with loud stertor and a mucopurulent or blood-stained nasal discharge. A high incidence of the disease may occur on some farms and in particular areas. Other diseases with similar clinical profiles include nasal obstruction associated with the blood fluke Schistosoma nasalis and chronic allergic rhinitis.1 REFERENCES

Diagnostic confirmation Response to treatment if no eggs/larvae in feces. Treatment Eprinomectin, ivermectin, fenbendazole (elevated dose); mebendazole over 5 days for donkeys. Control Avoid grazing donkeys and horses on same pasture.

ETIOLOGY Lungworm disease in horses is associated with the nematode parasite Dictyocaulus arnfieldi.

LIFE CYCLE

The life cycle of D. arnfieldi is direct and is almost identical to that of D. viviparus, except that the eggs do not hatch until shortly after they are passed in the feces.

EPIDEMIOLOGY

Infestations with D. arnfieldi are recorded more commonly in donkeys than in horses, and the former are considered to be the more normal host. Patent infections may persist in donkeys throughout their lives but in horses are generally confined to foals. These animals therefore provide the most important sources of pasture contamination. Nevertheless, a small proportion of infected adult horses shed low numbers of eggs, and this may be sufficient to perpetuate the life cycle even in the absence of donkeys and foals. As with D. viviparus, larvae can cross field boundaries by fungal transfer.

PATHOGENESIS

1. Nollet H, et al. Zoonoses Public Health. 2008;55:274. 2. Burgess HJ, et al. J Vet Diagn Invest. 2012;24:777. 3. Vilela R, et al. Revista Iberoamericana De Micologia. 2012;29:185. 4. Sudasinghe T, et al. Acta Trop. 2011;120:72. 5. Das S, et al. Med Mycol. 2011;49:311. 6. Peaty M. Vet Rec. 2007;160:883. 7. Leeming G, et al. Emerg Infec Dis. 2007;13:1377. 8. Leeming G, et al. Vet Rec. 2007;160:552. 9. Berrocal A, et al. Can Vet J. 2007;48:305.

Adult worms are found in the smaller bronchi, which they almost completely block. In adult horses however, few larvae reaching the lungs develop to this stage. Bronchioles in affected areas are surrounded by dense infiltrations of inflammatory cells, the epithelium becomes hyperplastic, and excessive mucus is produced. The consequent interference with airflow leads to patches of hyperinflation in the lung tissue.

LUNGWORM IN HORSES

CLINICAL FINDINGS

SYNOPSIS Etiology The nematode parasite Dictyocaulus arnfieldi. Epidemiology Infection is by ingestion of larvae on herbage; donkeys and foals shed most larvae, but adult horses can perpetuate life cycle. Signs Chronic cough in adult horses. Clinical pathology Eggs or larvae in feces (but often absent in affected adults); eosinophils in tracheal mucus. Lesions Discrete areas of hyperinflation in lung tissue.

Lungworm disease in horses is characterized by a chronic cough. Experimental infections produce an afebrile condition with coughing, increased respiratory rates, and forced expiration being most intense during weeks 3 to 5 after infection. Thereafter the signs decrease in severity but coughing may persist for several months. Heavy infestations in donkeys do not cause clinical illness. Horse foals may also be symptomless, although some show clinical signs.

CLINICAL PATHOLOGY

Characteristic eggs may be found in the feces of a small proportion of cases. Eosinophils and sometimes eggs or larvae may be demonstrated in tracheal mucus.

1047

NECROPSY FINDINGS The most obvious lesions at necropsy are discrete patches of overinflation.

DIAGNOSTIC CONFIRMATION

D. arnfieldi eggs in fresh feces are oval, are thin shelled, and contain a larva. Because the eggs may have hatched before arrival at the laboratory, it is usual to harvest larvae with the Baermann technique. The larvae resemble those of D. viviparus, but the tail ends in a small spine. Because many clinical cases are nonpatent and because tracheal mucus is difficult to sample, confirmation of diagnosis is often dependent on response to treatment. DIFFERENTIAL DIAGNOSIS • Recurrent airway obstruction (heaves) • Pulmonary abscessation and pneumonia • Inflammatory airway disease

TREATMENT TREATMENT Eprinomectin (0.5 mg/kg, top.) (R2) Ivermectin (0.2 mg/kg SQ) (R2) Mebendazole (20 mg/kg, q1d for 5 days) (R3)

Eprinomectin, as a pour-on formulation (0.5 mg/kg), has 100% efficacy in eliminating fecal larvae in donkeys within 7 days after treatment.1 Ivermectin at the standard equine dose is highly effective against immature and mature stages. For donkeys, mebendazole may be used at 15 to 20 mg/kg daily for 5 days, but this should not be attempted within the first 4 months of pregnancy.

CONTROL

Donkeys and horses should not be grazed on the same pasture. If this is impossible, the former should be treated regularly for lungworm. If there is a problem in a closed herd of adult horses, individuals with patent infection can be identified by fecal screening and treated. REFERENCE

1. Veneziano V, et al. Vet J. 2011;190:414.

Diseases of the Swine Respiratory Tract PROGRESSIVE ATROPHIC RHINITIS (CONCHAL ATROPHY OF SWINE) Atrophic rhinitis is a disease affecting primarily young pigs but causing anatomic lesions that may persist for life. The term nonprogressive atrophic rhinitis is used for the

1048

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

slight to severe rhinitis and usually transient atrophy of the conchal bones (formerly called the turbinates) in which no toxigenic P. multocida are found, when there are no clinical signs and no obvious growth retardation. This mild form is probably as a result of infection with Bordetella bronchiseptica (BB) or nontoxigenic P. multocida (PM). The term progressive atrophic rhinitis is proposed for the infection with toxigenic P. multocida (PM) (capsular serotype D and A strains) characterized by shortening or distortion of the snout, sneezing, nasal discharge, and epistaxis. Progressive atrophic rhinitis is often accompanied by reduced growth rates in severe cases. The organism is a zoonosis,1 but this is rarely from the pig,2 although pig farmers often have PM in their nasal cavities.3 SYNOPSIS Etiology Toxigenic strains of Bordetella bronchiseptica and Pasteurella multocida Epidemiology Young growing pigs. High percentage of pigs reared under intensive conditions may have some degree of atrophic rhinitis. Infection is widespread and transmitted by carrier sow to piglet. Housing and ventilation risk factors. Immunity develops in herd. Major economic importance because may affect growth rate and predispose to pneumonia. Signs Initially sneezing when piglets 3 to 9 weeks of age. Nasal discharge. Deformity of face with nasal bones (twisted snout). Growth rate may be decreased. Clinical pathology Culture organism from nasal swabs; polymerase chain reaction (PCR) Lesions Varying degrees of severity of atrophic rhinitis. Diagnostic confirmation Necropsy examinations of snouts. Differential diagnosis list • Inclusion-body rhinitis • Necrotic rhinitis • Inherited prognathia Treatment Antimicrobials in early stages; nothing later. Control Eliminating toxigenic strains of P. multocida. Depopulation and repopulation. Reduction of infection. Mass medication. Medicated early weaning. Vaccination.

ETIOLOGY Infection of the nasal cavities with BB followed by toxigenic strains of PM—primarily capsular type D and occasionally type A— results in progressive turbinate atrophy. PM has four subspecies (multocida, septica, gallicida, and tigris), but multocida is usually isolated from pigs4. There are five capsular serotypes (A-F) of PM. PM type A strains were formerly thought to be associated

entirely with lung infections but there is increasing evidence that some strains of PM type A are toxin producers and may be involved in atrophic rhinitis. Toxin production appears to be independent of serotype. The strains of PM isolated from the lungs are usually nontoxigenic and of capsular type A but a small proportion are toxigenic and are capsular type D. Serotype B is probably the most common one associated with septicemic pasteurellosis. The majority of cases of progressive atrophic rhinitis were associated with toxAcontaining capsular type D strains. Somatic antigens reflecting differences in lipopolysaccharides have also been used, in addition to a variety of other techniques.5 The poultry type of analysis6 based on multilocus sequence typing will be adapted for use for the pig eventually. There may a limited genetic heterogeneity in both the healthy pig strains and the PAR strains.7

EPIDEMIOLOGY Occurrence Atrophic rhinitis occurs worldwide where pigs are reared under intensive conditions. It has, however, become much less important with the onset of vaccination, improvement in resistance by pig breeding companies, and general attention to the environment in the farrowing house. Some surveys have shown that 50% of finished pigs and sows at slaughter have lesions of atrophic rhinitis. The incidence of clinical disease varies from 5% to 30%, which in part depends on the method of detection of the gross lesions. Abattoir surveys of the snouts of slaughtered pigs indicate that the incidence of gross lesions ranges from 14% to 50%. However, the incidence of gross lesions in abattoir surveys is biased by the source of the pigs; the incidence may be low in pigs from herds that have attempted to control the disease and high in some commercial herds with no control program. In pigs slaughtered from pig testing stations the incidence of lesions may be uniform over a long period. The published data on the incidence of gross lesions are also variable because of the lack of a uniform method of evaluating and quantifying the lesions. The incidence and severity of the lesions may vary with the season and the type of facility in which pigs are reared. In a slaughter survey of the snouts and lungs of pigs from 21 pig herds over one winter and one summer, the lesions of atrophic rhinitis were more severe among pigs slaughtered in the summer, whereas lesions of pneumonia were more severe among pigs slaughtered in the winter. Lesions of atrophic rhinitis were also more severe in pigs farrowed in central, enclosed farrowing houses and finished in enclosed, mechanically ventilated buildings than in pigs farrowed individually in sow huts and finished on dirt lots. It is possible that the incidence and severity of the lesions

at slaughter may be a reflection of the condition of the housing facilities when the animals were piglets several months previously, but many other factors could have been involved. Prevalence of Infection B. bronchiseptica readily colonizes the ciliated mucosa of the respiratory tract of pigs and infection of the nasal cavities of pigs is present in almost every pig herd, with the prevalence of infection in pigs in commercial herds varying from 25% to 50%. Serologic surveys of individual herds have found that up to 90% of the pigs are positive, which indicates that there is no reliable correlation between the frequency of isolation of the organism and the percentage of animals with antibody. The prevalence of infection is just as high in specificpathogen-free herds as in nonspecificpathogen-free herds. The prevalence of infection of toxigenic PM type D is higher in herds with clinical disease. The organism can be present in 50% to 80% of weaned pigs in a herd with clinical disease in the finishing pigs. Toxigenic type D PM was first detected in New South Wales, Australia, in 1986; in all herds examined, the introduction of pigs from an infected herd in South Australia was associated with an increased risk of infection. Toxigenic PM type D has been isolated rarely from herds free of atrophic rhinitis. Whereas BB is eliminated from the respiratory tract of most infected pigs, leaving only a few infected at slaughter, PM often persists. Method of Transmission Direct contact and droplet infection are presumed to be the most likely methods of transmission. The reservoir of infection is the infected sow, and litters of piglets become infected at an early age. Colonization of the tonsil by PM in conventionally reared pigs is common. In the Netherlands, it has been recognized that infection is usually by one of four possibilities. These are artificial insemination centers, laborers, neighborhood infection by direct aerosol or indirect local contact, and the presence of carrier animals and birds. The infection is usually introduced into a herd by the purchase of infected pigs. Spread between piglets is probably enhanced after weaning when mixing of litters occurs, and 70% to 80% of a large weaned group may become infected. Infection persists for up to several weeks and months, followed by a gradual reduction in the intensity and rate of infection. In herds where BB is the initiating agent, up to 90% of pigs 4 to 10 weeks of age will have nasal infection, but this infection rate falls to approximately 15% by 12 months of age, and the proportion of carrier pigs within the breeding herd decreases with increasing age of sow. The

Diseases of the Swine Respiratory Tract

VetBooks.ir

prevalence of infection is also much higher during the period from October to March than at other times of the year, and the prevalence of serologically positive animals is highest from July to December. This is most probably a result of the winter housing conditions, with few air changes per hour, fluctuating temperatures, and high humidity. The epidemiology of toxigenic strains of PM as a causative agent of atrophic rhinitis is not as well understood. The organism colonizes the tonsils of clinically normal pigs. In contrast to BB, which is ubiquitous in pig herds, the toxigenic isolates of PM appear to be restricted to herds affected with progressive atrophic rhinitis. The organism is invariably present in herds with progressive atrophic rhinitis but may also be present in about 5% of the pigs in a herd with no clinical history of atrophic rhinitis. The main source of toxigenic isolates of PM for young pigs appears to be the pharyngeal tissues of the breeding stock. About 10% to 15% of sows in farrowing houses may be infected with toxigenic isolates, and piglets become infected within a week after birth. In contrast to BB, infection of piglets at 12 to 16 weeks of age with toxigenic PM will still result in varying degrees of severity of lesions. It is possible for growing pigs to develop lesions of atrophic rhinitis well beyond the age of 3 weeks if they are exposed to pigs affected with disease and infected with PM and BB. Risk Factors Animal Risk Factors The age at which piglets first become infected with BB has an important effect on the development of lesions. The most severe lesions occur in nonimmune animals infected during the first week of life. Animals infected at 4 weeks of age develop less severe lesions, whereas those infected at 10 weeks do not develop significant lesions. Immune Mechanisms The level of immunity in the young pigs will influence the level of infection and the incidence of clinical disease. Colostral immunity from sows serologically positive to BB is transferred to piglets and provides protection for 2 to 5 weeks. Clinical disease does not occur in piglets with high levels of passive antibody. Older pigs from 10 to 12 weeks of age may become infected but are less likely to develop severe turbinate atrophy and may develop inapparent infection and become carriers. Vaccination of the sow before parturition to increase colostral immunity or vaccination of the young pig will increase the rate of clearance of the organism from the nasal cavity and reduce the incidence of clinical disease. In chronically affected herds a level of immunity develops with increasing age of the breeding herd.

Pathogen Factors The virulence characteristics of BB and the toxigenic isolates of PM are important risk factors. Both organisms are required to produce lesions similar to the naturally occurring progressive disease. The virulence of BB is dependent on the ability to produce heavy, persistent colonization in the nasal cavity and the production of a heat-labile toxin. Bordetella spp. produce several virulence factors and toxins, which are regulated by a two-component sensory transduction system encoded by the bvg locus. These virulence factors include adhesins such as filamentous agglutinin, pertactin, and fimbriae; the adenylate cyclase–hemolysin toxin; and the dermonecrotic toxin. In cell cultures the dermonecrotic toxin stimulates DNA and protein synthesis and assembly of actin stress fibers while inhibiting cell division, resulting in polynucleation of cells. It mediates these through the modification and activation of the small guanosine 5′-triphosphate (GTP)binding protein Rho. There are both toxigenic and nontoxigenic strains of BB. Colonization of the nose was greater with the dermonecrotic-toxinpositive strains than with the dermonecrotictoxin-negative mutant strains. This was maintained for the first, second, and third weeks postinoculation, but by the fourth week the position had changed to the opposite. All dermonecrotic-toxin-positive pigs had pneumonia but the dermonecrotictoxin-negative animals were able to colonize the lung more freely. There is an outer membrane protein P68 perlactin (BB perlactin gene [prn]), an adhesin, that may play a part in the protective immunity and may be extremely variable. The most important experiment is one that shows that PM mutant strains without the capacity to produce PM type D toxin did not produce turbinate atrophy. Only certain porcine phase 1 cultures possess both properties. However, even the most virulent of 10 isolates of BB did not cause progressive turbinate atrophy or significant snout deformation in experimental infections. The severe lesions of atrophic rhinitis cannot be attributed to this organism alone. Experimental inoculation of specificpathogen-free or gnotobiotic pigs with the organism results in a nonprogressive moderately severe turbinate atrophy 2 to 4 weeks after infection, followed frequently by regeneration of the turbinates. These virulence characteristics of BB are consistent with the observations that in herds where the organism is common it can provoke sneezing and coughing but no evidence of clinical turbinate atrophy. Examination of the turbinates within 2 weeks after the sneezing will reveal some mild lesions, but no lesions will be evident when the pigs are examined at slaughter. It may be that the adhesins left over in the nasal cavity from an infection of BB are subsequently available for the attachment of other bacteria.

1049

Toxigenic isolates of PM colonize the nasal cavities, elaborate several toxins, and produce progressive lesions of the turbinate bones and snout. Toxigenic PM can colonize the upper respiratory tract of pigs, and the presence of the capsule is a virulence factor. The presence of BB can enhance the colonization of PM, particularly the toxigenic type D strains isolated from pigs. The cytotoxin of BB is required for optimum growth by toxigenic PM; other products of phase 1 BB growth assist colonization by PM, and the degree of atrophy of the turbinates in these mixed infections is related to the numbers of toxigenic PM in the nasal cavity. Severe turbinate damage and shortening of the snout can be reproduced in specific-pathogen-free and gnotobiotic pigs by combined infection with BB and certain strains of PM. Following experimental infection both organisms may persist in the nasal cavities for up to 64 days. The cell envelope proteins and lipopolysaccharides of PM strains associated with atrophic rhinitis have been characterized and compared. At least three protein patterns and six lipopolysaccharide patterns can be distinguished, which can be used to predict the pathogenic character of some of the strains. This will obviate the need to use the guineapig skin test to distinguish those strains that are associated with atrophic rhinitis and those that are not. The gene for the osteolytic toxin of PM has been cloned and expressed in E. coli; the protein expressed has been shown to have the same properties as the native toxin. The toxin is the main colonization factor produced by toxigenic strains of the organism and antitoxin made from the toxin is protective experimentally and cross-protective between toxins from different capsule types. The toxin can produce turbinate atrophy when injected intranasally and also when given intramuscularly, intraperitoneally, intravenously, or intradermally. Fingerprinting techniques have been used to show that outbreaks of atrophic rhinitis since 1985 in Australia have been associated primarily with a single strain of toxigenic type D PM. Environmental Factors The effects of housing, population density, and adequacy of ventilation on the prevalence of infection of BB and toxigenic isolates of PM and on the incidence and severity of atrophic rhinitis have not been examined in detail. Atmospheric ammonia, dust, and microbial concentrations in the farrowing house and dust in weaner barns have a significant role in the severity of atrophic rhinitis. The mean daily gain of gilts with atrophic rhinitis exposed to ammonia may be smaller than that of those not affected. Undocumented field observations suggest that the disease is more common and severe when pigs are confined, overcrowded, and housed in poorly ventilated unsanitary

1050

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

barns, all of which promote the spread of infection. There is no effect of high levels of ammonia on the severity of turbinate atrophy. It has been shown that high levels of ammonia have no effect on the disease progression of atrophic rhinitis and pneumonia but do enhance the colonization of the nasal turbinates by toxigenic PM. A recent experiment has shown that higher numbers of PM bacteria were isolated from the tonsil than the nasal membranes per gram of tissue. Aerial pollutants contribute to the severity of lesions associated with atrophic rhinitis by facilitating colonization of the upper respiratory tract by PM. Management factors such as confinement farrowing and the use of continual throughput farrowing houses and weaner houses are also considered to be important risk factors. Adverse climatic conditions (below thermoneutrality with drafty periods) can result in a lower amount of energy available for production because of increased maintenance requirements, which results in growth retardation associated with lowered feed intake. Economic Importance Historically, it was accepted as dogma that atrophic rhinitis was an important cause of economic loss in pig herds because of decreased growth rate, less-than-optimal feed efficiency, and the fact that it was a major risk factor in enzootic swine pneumonia. A number of field studies have found an association between atrophic rhinitis and reduced growth rate in some herds, whereas other observations were unable to show an association between the presence of the disease and growth rate. The lack of a standard system for evaluation of conchal lesions may be a factor in the variable results between observations. Some field studies have failed to show that the disease has an effect on growth rate in finishing pigs or that there is a cause and effect relationship between atrophic rhinitis and pneumonia. The presence of pneumonia in pigs from a test station reduced mean daily weight gains by 33% for each 10% of affected lung, but atrophic rhinitis did not affect daily gain and there was no association between the development of atrophic rhinitis and the development of pneumonia. Pigs vaccinated against BB had turbinate atrophy scores or mean daily gains no different from those of unvaccinated pigs. In another study there was a low positive correlation between the herd mean turbinate atrophy score and the herd mean percentage pneumonia score. A recent report from Illinois indicates that the prevalence of clinical atrophic rhinitis in farrow-to-finish herds ranged from 0% to 20%, and in pigs from those herds examined at the abattoir the incidence of turbinate lesions ranged from 5 to 92%. In some of the herds the mean daily weight gain was 15% to

18% higher than in herds where pigs had severe turbinate lesions. In an Australian report there was no correlation between the severity of atrophic rhinitis and growth rate or back-fat thickness. In one study of three commercial pig herds, the snouts and lungs of individual pigs were examined and scored at slaughter, and the results were correlated with growth indicators for each pig (average daily gain during the growing and finishing phases, and days to reach market). Scores for lung lesions were also correlated to scores for snout lesions. Contrary to findings in many other studies, pigs that reached market weight at the youngest age did not have the lowest score for lung lesions, nor the lowest grade for snout lesions, nor the least extensive or severe lesions. It was concluded that lung lesions and grades for snout lesions in pigs at slaughter are not valid indicators for determining the economic effect of either pneumonia or atrophic rhinitis on growth performance of pigs.

PATHOGENESIS

Following infection of the nasal cavity, BB becomes closely associated with the ciliated epithelium of the respiratory tract. It can bind to respiratory tract mucus. The organism produces a heat-labile toxin that results in a nonprogressive, moderately severe turbinate atrophy that is apparent within 2 to 4 weeks after infection, followed frequently by regeneration of the conchae. There is, initially, ciliary loss and ciliary stasis, followed by reduction in mucociliary clearance, followed by hyperplasia and metaplasia of the nasal epithelium, fibrosis in the lamina propria, and resorption and replacement fibrosis of the osseous core. Experimental infection with BB alone does not result in severe persistent conchal atrophy or twisting or shortening of the snout. The strains of BB that produce cytotoxin may predispose to the colonization of PM in the nasal cavities. The preferred habitat of PM appears to be the tonsillar crypt, but following damage by BB, it can inhabit the epithelium of the URT. Infection and colonization of the nasal cavities, particularly the mucus, with the toxigenic strains of PM results in the elaboration of a toxin that causes progressive conchal atrophy. The toxin is thermolabile and dermonecrotic and is called the dermonecrotic toxin of PM. It interferes with G-protein and Rho-dependent signaling pathways in the cells. It is encoded by the toxA gene. The inoculation of a toxin from a toxigenic strain of type D PM into the nasal cavities of gnotobiotic pigs results in severe bilateral atrophy of the conchae. Atrophy of the ventral conchae can be produced experimentally with pathogenic BB in piglets at 6 weeks of age and with toxigenic PM strains in piglets as old as 16 weeks of age. The toxin enhances osteoclastic resorption and impairs osteoblastic synthesis of the

conchal osseous core; irreversible changes can occur within a few days. The toxin is a one chain toxin of 1285 amino acids, and different domains of the toxin are involved in cell uptake and intracellular activities. The toxin is able to subvert cell cycle progression and cell–cell signaling systems in osteoblasts and osteoclasts. The toxin is the sole agent responsible for the conchal atrophy, and the effect appears to be related to the total exposure to the toxin; that is, it is dose dependent. The toxin PMT activates various heterotrimeric G proteins, which causes the deamidation of the alpha-subunits of the G proteins.8-11 More important, this also appears to have an immunomodulatory effect. There is an inverse relationship between the number of PM and the total concentration of immunoglobulin. This may in part be one of the reasons that local changes in the nose produce such adverse growth effects, and they may be a result of the fact that the PM type D toxin has in fact changed the immune functions and that the PM may have predisposed to many other agents. These authors’ conclusion is that PM significantly suppresses the antigen-specific IgG immune responses of pigs to parenteral antigen challenge. The epithelium and the submucosa undergo secondary atrophy, and the conchae may disappear almost completely within 10 to 14 days. These lesions can persist until the animal is 90 kg in body weight. The conchal atrophy is not accompanied by an inflammatory reaction. The effect of the PM toxin is restricted to the nasal cavity; this is supported by the intriguing observation that the parenteral injection of the toxin into gnotobiotic piglets results in turbinate lesions and shortening and twisting of the snout. The parenteral injection of the dermonecrotoxin of PM capsular type D into specific-pathogen-free adult pigs will result in moderate conchal atrophy. In piglets 7 days of age, the intramuscular injection of the purified dermonecrotoxin will result in severe atrophy of the conchae. The culture filtrate of a nonatrophic-rhinitis pathogenic PM will not cause lesions after intramuscular injection. The disappearance of the conchae and the involvement of the bones of the face lead to deformity of the facial bones with the appearance of dishing and bulging of the face and, if the lesion is unilateral, to lateral deviation of the snout. The effect on growth rate, if any, may be attributable to the chronic irritation and interference with prehension. Experimentally, atrophic rhinitis suppressed the health of pigs, reducing their activity and feed intake. Experimentally, parenteral injections of the toxin decrease physeal area and reduce chondrocyte proliferation in long bones, in addition to conchal atrophy. Reliable experimental models of atrophic rhinitis in gnotobiotic pigs are now available and are useful for studying the pathogenesis of the disease and testing vaccine strategies.

Diseases of the Swine Respiratory Tract

VetBooks.ir

A sterile sonicate of a toxigenic strain of BB is instilled into the nasal cavities of piglets at 5 days of age followed by intranasal inoculation of toxigenic strains of PM at 7 days of age. The toxin can also affect the liver and urinary tract and decrease the physeal area in the long bones.

CLINICAL FINDINGS

The clinical findings of atrophic rhinitis depend on the stage of the lesions. In acute cases in piglets 3 to 9 weeks of age, irritation of the nasal mucosa causes sneezing, some coughing, small amounts of serous or mucopurulent nasal discharge, and transient unilateral or bilateral epistaxis. The frequency of sneezing may be a measure of the incidence and severity of the disease. In piglets born from sows vaccinated with BB and PM vaccine before farrowing, followed by two vaccinations within 3 weeks of age, the frequency of sneezing at 3 to 9 weeks of age was much less than in piglets given only BB vaccine. There may be rubbing of the nose against objects or on the ground. A watery ocular discharge usually accompanies this and may result in the appearance of dried streaks of dirt below the medial canthus of the eyes. There may be a decrease in growth rate. In infection with BB these clinical signs will disappear spontaneously in a few weeks, when the pigs will appear normal. In severe cases, respiratory obstruction may increase to the point of dyspnea and cyanosis, and sucking pigs may have great difficulty in nursing. The nasal secretions become thicker and nasal bleeding may also occur. In the more chronic stages, inspissated material may be expelled during paroxysms of sneezing. During this chronic stage, there is often pronounced deformity of the face as a result of arrested development of the bones, especially the conchae, and the accumulation of necrotic material in the nasal cavities. The nasal bones and premaxillae turn upward and interfere with approximation of the incisor and, to a lesser extent, the molar teeth. There are varying degrees of brachygnathia superior and protrusion of the lower incisor teeth. Prehension and mastication become difficult, with a resulting loss of body condition. Facial distortion in the final stages takes the form of severe “dishing” of the face with wrinkling of the overlying skin. If the condition is unilateral, the upper jaw may be twisted to one side. These visible facial deformities develop most commonly in pigs 8 to 10 weeks old within 3 to 4 weeks after infection, but they may occur in younger pigs. The most serious effects of the advanced disease are depression of growth rate and unthriftiness. The appetite may be unaffected, but much feed is lost by spillage, and feed efficiency may be reduced in some instances.

CLINICAL PATHOLOGY Culture and Detection of Bacteria It is important to be able to detect infected animals in a herd, especially the carrier animal. Nasal swabs are used to detect the bacteria and to determine their drug sensitivity. The collection of the nasal swabs must be done carefully and requires a special transport medium to ensure a high recovery rate. A sampling technique and a special culture medium to facilitate the isolation and recognition of BB are described. The external nares are cleaned with alcohol, and a cotton-tipped flexible wire is pushed into the nasal cavity (of each side in turn) until it reaches a point midway between the nostril and the level of the medial canthus of the eye. On removal, the cotton tip is cut off into 0.5 mL of an ice-cold sterile transport medium comprising phosphatebuffered saline (PBS, pH 7.3) with fetal calf serum (5% v/v). The samples are then placed on special media, preferably within 4 hours. Normally the organism grows well on conventional culture media, especially when younger pigs are sampled. However, in the carrier pig the organism may be sparse, and the selective medium is recommended. The nasal culturing procedure has been used as an aid in the control of atrophic rhinitis associated with BB. A series of three nasal swabs from each animal is considered to be about 77% efficient in detecting infected animals for possible culling and elimination from the herd. However, in some studies there may be no marked difference in the prevalence of BB or PM in pig herds with or without clinical atrophic rhinitis. Toxigenic PM grow readily in the laboratory but are difficult to isolate from nasal swabs because they are frequently overgrown by commensal flora. Selective laboratory media containing antimicrobial agents have been developed to promote the isolation of PM from nasal swabs. Inoculation of cotton swabs to selective medium on the same day as the sampling provides the best isolation of toxigenic PM. Immersion of pigs at slaughter in the scalding tank can result in a marked reduction in the isolation of toxigenic PM. A cell culture assay using embryonic bovine lung cell cultures is available and is a sensitive in vitro test for the differentiation of toxigenic from nontoxigenic isolates of PM. This test can replace the lethal tests in mice or the dermonecrotic tests in guinea pigs. Serology Agglutination tests and an ELISA test are available for the detection of pigs infected with BB, especially carrier animals. Serology is of value in the assessment of the response of pigs vaccinated with the BB vaccines. There are currently no reliable serologic tests for Pasteurellae.

1051

Antigen Detection A PCR method originally described in 1996 for the enhanced detection of toxigenic PM directly from nasal swabs has been described and upgraded. This was shown to be 10 times more sensitive than PM type D toxin (PMT) ELISA and 5 times more sensitive than clinical bacteriology with subsequent use of PMT ELISA. A nested PCR has also been described. Similarly, a PCR method for the detection of BB has been described that produces 78% more positives than culture, particularly with swabs with a high mixed bacterial load. Recently a nested-PCR has been described that was reported to be more specific and sensitive than the other PCR methods previously described. It does not require culture, it is less laborious, and the results can be provided within 24 hours. The authors concluded that this test was suitable for breeding company evaluations and for eradication schemes. Radiography Some aids to the clinical diagnosis have been examined but are not highly accurate. Radiography of the nose is not reliable in detecting the severity of conchal atrophy.

NECROPSY FINDINGS

The typical lesions of atrophic rhinitis are restricted to the nasal cavities, although concurrent diseases, especially virus pneumonia of pigs, may produce lesions elsewhere. In the early stages there is acute inflammation, sometimes with the accumulation of pus, but in the later stages, there is evidence only of atrophy of the mucosa and decalcification and atrophy of the conchae and ethmoid bones, which may have completely disappeared in severe cases. The inflammatory and atrophic processes may extend to involve the facial sinuses. There is no evidence of interference with the vascular supply to the affected bones. The changes in the nasal cavities are most readily seen if the head is split in the sagittal plane but for accurate diagnosis the degree of conchal symmetry, volume, and atrophy and medial septum deviation should be assessed by inspection of a vertical cross-section of the skull made at the level of the second premolar tooth. The clinical diagnosis is confirmed and the severity of the lesions is assessed by the postmortem examination of a cross-section of the snout. The snout must be sectioned at the level of the second premolar tooth because the size of the conchal bone reduces anteriorly and may give a false-positive result if the section is taken too far forward. Quantification of the severity of the lesions has been of value for monitoring the incidence and severity of the disease in a herd. Several systems have been used for grading the severity of lesions of the snout. Most of them have used a subjective visual scoring system in which snouts are grade 0 (complete normality) to 5 (complete conchal atrophy).

1052

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Reasonable agreement among observers recording morphologic changes of nasal conchae is achievable with some training. The standards for each grade are as follows: • Grade 0: No deviation from absolute normality, with nasal septum straight and conchae symmetric and filling nasal cavities. • Grade 1: Slight irregularity, asymmetry, or distortion of the nasal structures without atrophy. • Grade 2: Marked distortion of nasal structure but without marked atrophy. • Grade 3: Definite atrophy of the conchae with or without distortion. • Grade 4: More severe atrophy with severe atrophy of one or more conchae. • Grade 5: Very severe atrophy in which all conchae have virtually disappeared. Such a discontinuous grading system does not provide a direct quantitative relationship. Regular examination of the snouts from heads of pigs sent to slaughter can be used to assess the level of conchal atrophy in the herd. Morphometric methods, using either point counting or semiautomated planimetry applied to photographic or impression prints of sections of the snout to measure the extent of conchal atrophy on a continuous scale as a morphometric index, are now available. Cross-sections of the snout are photographed or used to make impression prints, which are then measured. A morphometric index is determined, which is the ratio of free space to total cross-sectional area of the nasal cavity. The system correlates well with the visual grading system of 0 to 5 but is labor-intensive and relatively expensive. The conchal perimeter ratio may be a more reliable morphometric measure of atrophic rhinitis and also provides parametric data suitable for quantitative analysis. A morphometric analysis using conchal area ratio is the best method for quantifying gross morphologic turbinate changes. Descriptions of the methods for making snout impressions are available. Computed tomography has been described. A major limitation of the grading system is that conchal atrophy occurs as a continuous spectrum, and it is difficult to decide, for example, if a pig with a grade 3 lesion represents the more severe manifestation of BB infection, which may not progress further, or an early manifestation of infection with toxigenic PM, which could develop into a severe herd problem. Histologically, the lesions vary according to the stage of the disease; initially there is a neutrophilic infiltrate followed by more chronic mononuclear cell infiltration. The conchal bones are eroded by osteoclasts, and new bone formation is reduced with degeneration dystrophy and reparative processes.

Samples for Confirmation of Diagnosis • Bacteriology—nasal swabs are not as good as tonsil swabs but are easier to obtain. The highest isolation rates are achieved with Knight medium or KPMD. Conventional biochemistry can then be used to identify.12 • Histology—formalin-fixed crosssection of snout at level of second premolar • Antigen detection—nasal swabs. ELISAs based on the use of PMTspecific monoclonal AB are rapid, sensitive and specific. The kmt1 gene has been used as a target for the loop-mediated isothermal amplification method.13 Diagnostic tests have been reviewed.14 Computer tomography can be helpful.15 DIFFERENTIAL DIAGNOSIS The occurrence of sneezing in the early stages and of facial deformity in the later stages are characteristic of this disease. Diagnosis depends on clinical signs, pathology, and demonstration of PM and its toxin. Inclusion-body rhinitis as a result of a cytomegalovirus is a common infection in young piglets in which there is sneezing and conjunctivitis. However, by itself it does not progress to produce turbinate atrophy and facial distortion. Under good hygienic conditions the course of the disease is about 2 weeks, and the economic effects are minimal. In the early acute stages, atrophic rhinitis may be mistaken for swine influenza, which, however, usually occurs as an outbreak affecting older pigs and accompanied by a severe systemic reaction without subsequent involvement of facial bones. Necrotic rhinitis is manifested by external lesions affecting the face, and virus pneumonia of pigs is characterized by coughing rather than sneezing. The inherited prognathic jaw of some breeds of pigs has been mistaken for the chronic stage of atrophic rhinitis; protrusion of the lower jaw is quite common in adult intensively housed pigs and has been attributed to behavioral problems of pushing the snout against fixed equipment such as bars and nipple drinkers.

TREATMENT Treatment early in the course of the disease will reduce the severity of its effects, but it is of little value in chronically affected pigs, and these pigs are best culled at an early age because of their persistent poor growth rate and high food conversion. Tylosin at 20 mg/kg BW, oxytetracycline at 20 mg/kg BW, or trimethoprim–sulfadoxine (40 mg/200 mg/mL) at 0.1 mL/kg BW may be given parenterally, or the creep feed may be medicated with sulfamethazine and/

or tylosin at 200 and 100 mg/kg of feed respectively. Parenteral injections need to be repeated every 3 to 7 days for at least three injections, and feed medication should be given for 3 to 5 weeks. The problem with early creep medication is in obtaining adequate intakes of the antibacterial. This is seldom achieved before 2 weeks of age, and parenteral antibiotics may be required if significant infection occurs before this stage. The parenteral administration of antimicrobial agents to individual piglets at - to 7-day intervals beginning at 3 days of age for a total of three to five injections per piglet has been recommended for the treatment and control of atrophic rhinitis. However, in a large herd such a treatment regimen would be a major task, and until a cost–benefit analysis indicates a beneficial effect over other methods, we cannot recommend such a practice. The treatment of experimental BB infection in young pigs has been successful with the use of trimethoprim–sulfadiazine in the drinking water at levels of 13.3 and 77.6 µg/ mL respectively, for 3 weeks. This method would remove the necessity to inject pigs repeatedly. Tilmicosin has proved useful; fed continuously over 6 weeks at concentrations of 200 g per ton of feed, it controlled transmission of atrophic rhinitis, weight gains were positively affected, and fewer nasal swabs were positive for PM at the end of the study period. A resistance to some antibiotics has recently been reported.12,16

CONTROL

Effective control depends on developing methods of eliminating or controlling the prevalence of toxigenic isolates of PM, which cause progressive atrophic rhinitis if they become established in the nasal cavity. Previous infection of the nasal cavity with BB may enhance the establishment of toxigenic PM and result in progressive atrophic rhinitis. Although there is considerable information available on the ecology of BB and the methods by which it might be eliminated or controlled in a herd, there is little documented information available on methods that can be used for control of the toxigenic isolates of PM associated with atrophic rhinitis. Control of atrophic rhinitis can be attempted in at least four ways: • Total eradication • Reduction of infection pressure • Mass medication with antimicrobials to reduce the severity and adverse effects of infection • Vaccination Regardless of the method employed, any effective control program must have a system for monitoring the incidence of clinical disease in the herd and the incidence and severity of conchal lesions of the pigs sent to slaughter. Accurate and reliable methods for monitoring clinical disease are not available,

Diseases of the Swine Respiratory Tract

VetBooks.ir

but the incidence of acute rhinitis and facial deformities could be recorded regularly. At slaughter, snouts can be examined for lesions of conchal atrophy and for assessing a mean snout score for each group of pigs slaughtered. Eradication Total eradication can only be achieved with confidence by complete depopulation for a 4-week period and repopulation with primary or purchased specific-pathogen-free stock. This approach has the added advantage of also eliminating enzootic pneumonia, which may be a significant contributing factor to the economic importance of this disease. However, this method of control is extremely costly, and the economic importance of the disease would need to be carefully evaluated in relation to this cost before this method was instituted. Other techniques of obtaining pigs free of atrophic rhinitis, such as the isolated farrowing of older and presumed noncarrier sows with subsequent clinical and postmortem examinations of a proportion of the litters, have had a significant failure rate in the field and are not recommended. Eradication by repopulation with cesarean-derived stock may be essential in breeding nucleus herds where a high generation turnover results in a low herd sow age and a low herd level of immunity. The breakdown rate of herds established by this method can be significant, presumably because the initiating organisms are not solely confined to pigs. A pilot control scheme was initiated in Britain in which a herd had to meet the following conditions: • It must be inspected by a veterinarian every 6 months over a period of 2 years, over which time there must be no clinical evidence of atrophic rhinitis. • The herd owner must certify that atrophic rhinitis has not been suspected over the same time period. • Cross-sections of snouts taken from at least 30% of marketed pigs must be examined regularly by a veterinarian, and over a 2-year probationary period the average six-month snout score must not exceed 0.5. • There must be no vaccination or treatment for atrophic rhinitis. • New breeding stock can be introduced only from other qualified herds or herds derived by hysterectomy, artificial insemination, or embryo transfer techniques. Over a 5-year period 45 herds qualified at some stage, and 34 were still qualified at the end of 5 years. As of 1988, some herds had exceeded the snout score limit of 0.5, with their average scores increasing to 2.24. In these herds, there was no clinical, epidemiologic, or bacteriologic evidence that they

were at risk of developing severe atrophic rhinitis. It is suggested that the higher scores were associated with a group of recurrent husbandry factors, especially overstocking and unsatisfactory conditions in the weaner barns. These increased scores suggested the possibility that the upper limit for the snout scores in qualifying herds could be raised and allow bacteriologic testing to be confined to more doubtful herds. Eradication in the Netherlands was based on the fact that they thought that there were four main possibilities for the spread of toxigenic PM: artificial insemination centers, laborers, neighborhood infection either by aerosol or by local spread, and carrier animals or birds. They assumed that most herds were closed or buying certified stock and that the major source of infection was therefore the boar. In this study they tested boars; in herds with less than 50 boars they tested all, and in those with more than 50 they tested 50 as the minimum. They took nasal and tonsil samples, which were placed in cold transport medium and sent to the laboratory within 24 hours under cooled conditions for overnight culture followed by PCR. Reduction of Infection Reduction of infection pressure can be attempted. Infection of piglets occurs primarily either from carrier sows or from other infected piglets in the immediate environment and severe atrophic rhinitis generally results from infection of piglets under 3 weeks of age. If these factors can be minimized, the incidence and severity of the disease can be reduced. An all-in, all-out pig flow is one of the most effective methods of control of atrophic rhinitis. Changing to an all-in, all-out pig flow from continuous flow management can improve snout scores by 50%, lung scores by 55%, average daily gain by 0.14 lb, and days to market by 13 days. Because severe lesions depend on infection of the piglet under 3 weeks of age, every attempt should be made to minimize the severity of the challenge to young piglets. It is a common observation that the effects of atrophic rhinitis are minimal under good systems of management and adequate ventilation, nondusty conditions, and good hygiene. The use of continual-throughput farrowing houses and weaner houses allows a buildup of infection with the presence of actively infected pigs that can provide a high infection pressure on piglets born into or introduced into these areas. The use of all-in, all-out systems of management in these areas is recommended, and young piglets should be kept in a separate area from older pigs. Mass Medication The prophylactic use of antimicrobials is frequently employed to reduce the incidence of the disease within the herd. Antimicrobials are used both within the breeding herd to

1053

reduce the prevalence of carriers and in young suckling and weaner pigs to reduce the severity of the infection. The medication is begun about 2 weeks before farrowing, continued throughout lactation, and incorporated in the creep feed for the sucking pigs and the starter feeds for the weaned pigs. In this way there is continuous medication of the sow and the piglets during the most susceptible period. For the breeding herd, sulfamethazine at levels of 450 to 1000 mg/kg feed, with the higher levels being given to dry sows on restricted feeding, has been recommended. Sulfonamide resistance has proved a problem in some countries but beneficial results may still be achieved with these levels. It is recommended that medication be continued for a 4- to 6-week period. Carbadox at a level of 55 ppm in combination with sulfamethazine at 110 ppm is reported to be effective in clearing experimentally induced BB infection, and when used alone improved growth rate and feed efficiency in pigs with naturally occurring atrophic rhinitis. In the starter period, carbadox fed alone or in combination with sulfamethazine improved average daily gain in piglets from herds with naturally occurring atrophic rhinitis. Use of the medication, however, did not result in a reduction of mean nasal lesion scores as a result of atrophic rhinitis. Sulfamethazine at 110 mg/kg of feed is more effective than sulfathiazole at the same concentration for the control of experimentally induced atrophic rhinitis attributable to BB. Sulfamethazine may also be incorporated in creep rations, and the use of tetracyclines (200 mg/kg), tylosin (50-100 mg/kg), and penicillin (200 mg/kg) has also been suggested. Medicated early weaning is recommended to obtain pigs free from pathogens, including BB that are endemic in the herd of origin. The sows are fed medicated feed from 5 days before to 5 days after weaning, and the piglets are dosed from birth to 10 days of age. Vaccination There has been considerable interest in the development of vaccines for the control and prevention of atrophic rhinitis attributable to BB. Inactivated vaccines have been used to vaccinate the pregnant sow 4 to 6 weeks before farrowing; in some cases, this is followed by vaccination of the piglets at 7 and 28 days of age. In general, the use of the vaccine in pregnant sows in herds where the disease has been endemic has reduced the incidence of clinical atrophic rhinitis. However, the results from one study to another have been highly variable. Vaccination of the pregnant sow results in an increase in colostral antibody titer, which does improve the clearance rate of BB in the piglets. However, it has been difficult to evaluate the efficacy of the BB used alone because the conchal atrophy associated with infection of piglets with BB experimentally or naturally heals and regenerates completely

1054

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

when they are reared to about 70 to 90 kg BW in good housing conditions. Vaccination with both components (BB and PM) in a vaccine reduces lesions considerably compared with a placebo and a group with only PM type D toxin in the vaccine, but neither vaccine eliminated toxigenic PM from the upper respiratory tract. Experimentally, piglets born from sows vaccinated with PM are protected from a challenge with atrophic rhinitis toxin. This indicates that artificial immunization for atrophic rhinitis should be possible. Vaccination of sows at least three times before farrowing for the first time and during each subsequent pregnancy with a vaccine containing BB and PM was highly successful in reducing the incidence of atrophic rhinitis in the pigs. The incidence in affected herds was reduced from 7.5% to about 2%. Experimentally, the vaccine provides good protection against challenge in piglets from vaccinated sows. A recombinant PM toxin derivative vaccine given to gilts 4 to 5 weeks before farrowing and again 2 to 3 weeks later provided excellent protection in their piglets against experimental challenge with BB and toxigenic PM. This indicates the excellent immunoprotective properties of the nontoxic derivative of the PM toxin. In five field trials, a single-component vaccine containing a nontoxic but highly immunogenic protein, as the antigen, provided much better protection than the control vaccine containing killed PM and killed BB. Experimental infection and vaccination of pregnant minimum-disease sows with BB resulted in much higher agglutinins in serum and colostrum than in sows only vaccinated or control animals, and the piglets were provided with protection against experimental disease. Vaccination of pregnant gilts with purified inactivated PM toxin resulted in a high degree of protection of their progeny against progressive atrophic rhinitis. A new vaccine has been described using a truncated PM type D toxin that is immunogenic and nontoxic, a toxoid for BB, and an adjuvant. Sows were vaccinated at 8 to 6 weeks and 4 to 2 weeks before farrowing. The vaccinated animals had fewer organisms. FURTHER READING Horiguchi Y. Swine atrophic rhinitis caused by Pasteurella multocida and Bordetella dermonecrotic toxin. Curr Top Microbiol Immunol. 2012;36:1113-1129.

REFERENCES

1. Wilkie W, et al. Curr Top Microbiol Immunol. 2012;361:1. 2. Migliore E, et al. Adv Med Sci. 2009;54:109. 3. Marois C, et al. J Appl Microbiol. 2009;107:1830. 4. Varga Z, et al. Acta Vet Hung. 2007;55:425. 5. Dziva E, et al. Vet Microbiol. 2008;128:1. 6. Subaaharan S. Vet Microbiol. 2010;141:354. 7. Bethe A, et al. Vet Microbiol. 2009;139:97.

8. Orth JH. Proc Natl Acad Sci United States. 2009;106:7179. 9. Orth JH. Curr Top Microbiol Immunol. 2012;361:73. 10. Orth JH, et al. FASEB J. 2013;27:832. 11. Bergmann S, et al. Infect Immun. 2013;81:2459. 12. Lizarazo YA, et al. Am J Vet Res. 2006;67:663. 13. Sun D, et al. Vet Res Comm. 2010;34:649. 14. Stepniewska K, Markowska-Daniel I. Bull Vet Inst Pulawy. 2012;56:483. 15. Jablonski A, et al. Vet Rec. 2011;168:329. 16. Tang X, et al. J Clin Microbiol. 2009;47:951.

FACIAL NECROSIS (FACIAL PYEMIA) Facial necrosis (facial pyemia) was formerly called necrotic rhinitis or bullnose or paranasal abscessation and is often confused with atrophic rhinitis (AR). It occurs in growing pigs usually before 1 week of age and may occur in herds where AR is present and even in the same pig, but there appears to be no relationship between the two diseases. The diseases differ by the presence of oral and facial lesions. Necrotic ulcer in pigs may involve the mouth and face, but the lesions are erosive rather than necrotic. There are a variety of other conditions of the face of the young pig that can be confused. The common occurrence of Fusobacterium necrophorum in the lesions suggests that any injury to the face or nasal or oral cavities may lead to bacterial invasion, especially if the environment is dirty and heavily contaminated. The disease is now rarer following a general improvement in hygiene in piggeries but possibly also as a result of the declining occurrence of AR following vaccination and eradication of P. multocida toxigenic type D and much greater care in teeth clipping of the young pig. It is also associated with fighting in piglets trying to reach a teat, especially when milk is in short supply. The lesions develop as a necrotic cellulitis of the soft tissues of the nose and face but may spread to involve bone and produce osteomyelitis. Local swelling is obvious, and extensive lesions may interfere with respiration and mastication. The lesions may be ulcerated, crusty, and extensive. Depression of food intake and toxemia may result and poor growth, and some deaths result. Treatment by the local application of debridement, disinfection with substances such as chlorhexidine or iodophors and the use of antibiotic creams and parenteral antibacterial drugs, and the oral administration of sulphonamides is satisfactory in early cases. Oral dosing with sulphadimidine has been effective in young pigs. Improvement of sanitation, elimination of injuries, and disinfection of pens usually result in a reduction of incidence, and cross-fostering will reduce competition and fighting. FURTHER READING Done JT. Facial deformities of the pig. Vet Ann. 1977;17:96.

BORDETELLA RHINITIS Bordetella bronchiseptica (BB) is capable of causing two major disorders on its own. The first is Bordetella rhinitis, and the second Bordetella bronchitis. It is also capable of infecting man but the contribution of pig strains to human disease is unknown.

ETIOLOGY

It is a small, aerobic, gram-negative bacterium that produces a beta hemolytic 1- to 2-mm gray colony on some nutrient blood agars but is nonhemolytic on some enriched media. On MacConkey media it produces nonlactose fermenting colonies in 48 hours. Nearly all the strains express one of two antigenically distinct O-antigen serotypes (O1/ O2) that are not cross-reactive.1 Variation in virulence can result from strain variation2,3 and may be related to different phylogenetic lineages.4

EPIDEMIOLOGY

The bacterium is often isolated from healthy animals.5 Carrier animals usually introduce it to a herd. Strains from other animals (dogs, rodents, etc.) are not so likely to colonize the pig because only a few strains occur in the pig, and these tend to be different from other species. Spread is by aerosol from sneezing and through direct and indirect contact. Infection usually occurs early in life and what happens then depends usually on the state of immunity. Maternal antibody usually lasts long enough to cover the establishment of infection and prevents pathology but does not lead to removal of the agent. Cross-fostering; multiple ages in the same house; multisourcing to a nursery or finishing house; poor ventilation and environmental control and, in particular, lack of an all-in, all-out policy followed by effective cleaning, disinfection, and drying policy are conducive to the spread of the condition.

PATHOGENESIS

Bordetella is a complicated organism with several virulence factors. It exists in four colony phases. Expression of the virulence genes requires cooperation of the BvgAS (virulence genes system).6 Phase I colonies contain fully virulent organisms (Bvg +) expressing genes for flagellae (fla), the mannose-resistant filamentous hemagglutin,7,8 and the outer membrane protein pertactin (PN), all of which are involved in adhesion. Other factors include a hemolysin that is adenylate cyclise, a cytotoxin, an osteocytic toxin, and the dermonecrotic toxin (dnt). The adenylate cyclise may modulate cytokine production in dendritic cells and alter immunomodulatory function.9 The tracheal cytotoxin is likely to act on the cilia and cause ciliostasis. The Bvg + organisms also possess the bfrZ gene for the exogenous ferric siderophore receptor, which

Diseases of the Swine Respiratory Tract

VetBooks.ir

is essential because BB has huge requirements for iron. Phases II and III do not have all these. Reversion to phase I only takes place in vivo. The organisms also differ between strains in the presence of genes for flagellae and fimbriae. The organisms colonize the cilia of the URT and then proliferate, and then the cilia are lost as the organisms increase further in number. Pertactin may be required for this.9 Toxic substances then diffuse from the BB into the epithelium and below and damage the osteoblasts. Mild turbinate atrophy may then begin but usually resolves by about 70 days postinfection. In the lung, BB causes a pneumonia similar to Mycoplasma hyopneumoniae (EP), and the organism lives in large numbers in the main bronchi (formerly called bronchitis X), where it may cause a mucopurulent tracheitis and bronchitis. The organism also enhances the ability of other organisms to colonize the respiratory tract, notably P. multocida,10 S. suis, and H. parasuis. In turn, PRRSV predisposes to infection with BB. Coinfection of BB with PRCV and SIV leads to a longer outbreak of more severe pneumonia.11

CLINICAL SIGNS

Clinical signs may be severe in newly established herds, rapidly expanding herds, or in herds with poor immunity or where there are immunosuppressive disorders. Normally, outbreaks of sneezing will occur in baby pigs. It may be paroxysmal or be accompanied by epistaxis. There may be tear staining. The signs of progressive atrophic rhinitis are rarely seen with just BB infection. In the pneumonic form there may be fever to 40°C (104 F), anorexia and loss of condition, and possibly a high mortality. It may cause a reduction in growth rate that may reach 20% to 30%. Coinfection with other agents contributes to an increased severity of signs, and respiratory viruses may favor the colonization by BB.12

PATHOLOGY

In an uncomplicated infection there is a mild catarrhal rhinitis. There may be some degree of conchal (turbinate) atrophy with deviation of the nasal septum, and excess mucus production. In the lung infection there may be consolidation of cranial and middle lobes of the lung. Histologically, the nasal epithelium is infiltrated with inflammatory cells, it sometimes shows mucous metaplasia, and there may be fibrosis that is almost pathognomonic for BB infection. In the lung there may be a catarrhal exudate with neutrophilic infiltration.

DIAGNOSIS

In early cases, severe sneezing and tear staining will be a good indication. Sneezing is

the method of clearing the nasal cavity of irritation (infection, noxious gases such as ammonia or heavy burdens of dust) and is the clinical sign indicating the nasal cavity is stressed. In early cases of bronchial infection there may be a cough, which indicates that the trachea, main-stem bronchi, and the major part of the bronchial tree are clogged with exudate that needs to be physically removed because the normal mucociliary clearance mechanism is overcome. In early infections BB can be isolated from the whole of the respiratory tract, but in chronic or recovered cases it may only be isolated from the nasal cavity (ethmoturbinates in particular). Nasal swabs using cotton tips can be collected, placed in transport media, and cultured on special media. At postmortem the BB can be grown on blood agar plates with 48 hours of incubation. PCR tests based on the dermonecrotic toxin have been used successfully13 and in multiplex PCRs with P. multocida. Antibody tests (agglutination and ELISAs) can also be used to assess the herd status.

IMMUNITY

There is an IgM immunity to the hemagglutinin within 7 days and IgG appears 4 to 5 weeks later. This immunity usually prevents turbinate atrophy and pneumonic damage. It is necessary for a good IgA response to clear the URT of infection,14 but vaccine protection is not as good as natural infection protection.15

TREATMENT

Parenteral treatment with almost all antibiotics is possible for severe acute case because in vitro sensitivity to most antibiotics is high. Only after this should treatment via water and food should be considered. BB are, however, largely resistant to Ceftiofur, and there is evidence that they are becoming more resistant to trimethoprim–sulphonamide combinations

CONTROL

Medication can be used to control the onset of the problem. Threatened pigs in a single airspace should all be given antibiotics in the feed after weaning (trimethoprim/sulphonamides at 30 mg/kg daily) or tetracyclines. Strategic medication using the same antibiotics, given parenterally, at 3, 10, and 21 days of age will also reduce the clinical signs. Medicated early weaning techniques and long-term treatment in the water for 28 days have also been used to eradicate the agent. Vaccination using formalin killed alum adjuvenated vaccines usually combined with P. multocida toxoid have been successfully

1055

used for a long time.16 They can be given to sows 6 and 2 weeks before farrowing to stimulate maternal antibody and to piglets at 7 and 28 days of age, but in this case they may be negated by maternal antibodies. All-in, all-out management, with good ventilation reduces the level of infection. Purchasing clean stock with a period of isolation and quarantine will also remove the infection, as will treating incoming stock. It is sensitive to several on-farm disinfectants.17 REFERENCES

1. Buboltz AM, et al. Infect Immun. 2009;77:3249. 2. Buboltz AM, et al. J Bacteriol. 2008;190:5502. 3. Buboltz AM, et al. Infect Immun. 2009;77:3969. 4. Cummings CA, et al. J Bacteriol. 2006;188:1775. 5. Palzer A, et al. Vet Rec. 2008;162:267. 6. Beier D, Gross R. Adv Exp Med Biol. 2008;631:149. 7. Irie Y, Yuk MH. FEMS Microbiol Lett. 2007;275:191. 8. Nicholson TL, et al. Infect Immun. 2009;77:2136. 9. Vojtova J, et al. Curr Opinion Microbiol. 2006;9:69. 10. Brockmeier SI, Register KB. Vet Microbiol. 2007;125:284. 11. Brockmeier SI, et al. Vet Micrbiol. 2008;128:36. 12. Loving CL, et al. Microb Pathog. 2010;49:237. 13. Register KB, De Jong KD. Vet Microbiol. 2006;117:201. 14. Wolfe DN, et al. Infect Immun. 2007;75:4416. 15. Gopinathan I, et al. Microbes Infect. 2007;9:442. 16. Hsuan SI, et al. Vaccine. 2009;27:2923. 17. Thomson JR, et al. Pig J. 2007;60:15.

PLEUROPNEUMONIA OF PIGS ASSOCIATED WITH ACTINOBACILLUS PLEUROPNEUMONIAE ETIOLOGY Actinobacillus pleuropneumoniae (APP), formerly known as Haemophilus pleuropneumoniae, is the causative organism of pleuropneumonia in pigs. Some strains require V factor (NAD) for growth (biotype I), but some strains do not require this factor (type II). It forms small translucent mucoid betahemolytic colonies around staphylococcal streaks on sheep blood agar. It is a small gram-negative, encapsulated rod. The organism causes severe, rapidly fatal fibrohemorrhagic and necrotizing pleuropneumonia. The survivors often have bacteria-laden sequestra in the lungs that are poorly penetrated by antibiotics but do act as sources of the organism for later outbreaks. It does not affect humans and has no public health significance. Recently a completely nonpathogenic species, A. porcitonsillarum, has been identified.

EPIDEMIOLOGY Occurrence It is widely distributed worldwide. The primary reservoir is domesticated pigs, but wild boar are also affected.1 The only natural host is the pig but it has been isolated from cattle, deer, lambs, and

1056

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

some rodents can be infected experimentally. It is probably not carried by birds and rodents. The diversity of strains isolated from healthy pigs could be higher than that of strains recovered from diseased pigs. It appears that few pigs are infected from their sows, and then the organism spreads after weaning as the maternal antibodies disappear. The disease occurs worldwide in growing pigs from 2 to 6 months of age, with rapid spread both within the initially affected group and subsequently to other older or younger pigs in a herd. There are probably large numbers in the nose of affected animals. Abattoir surveys have found that the lungs of pigs from about 50% of herds monitored for several months may have lesions attributable to APP. This chronic pleurisy is presumably associated with APP.2,3 Sero-epidemiologic surveys have found that pigs in 70% of herds may have antibodies to one or more of several recognized serotypes of the organism. The prevalence of infection continues to increase—presumably as a result of confinement rearing, crowding, inadequate ventilation, close contact, and commingling of pigs of various age groups. The incidence of clinical disease is much less than the prevalence of infection. In most countries, the more dense the pig population, the more prevalent the APP is likely to be, and this has been documented in Belgium. In the United States it may not be as important as in Eurasia, and it was not reported to be of major importance at the Iowa clinic.4 In Europe, it may have become more common in recent years. Most herds have one or more types but these are usually avirulent. In some countries there may have been a shift from virulent to avirulent serotypes. In Canada, over 75% of the pigs were positive for APP in the upper respiratory tract.5 There is a relative homogeneity within a particular serotype.6 A strain may be virulent in one country but not in another depending on the genetic makeup determining the presence of virulence factors. Morbidity and Case Fatality The morbidity rate can exceed 50%, and the mortality may vary from 1% to 10%. Methods of Transmission Transmission is usually by pig to pig contact or more correctly nose-to-nose contact. Aerosol droplets only carry over short distances before they are precipitated. In Denmark it was suggested that the paramount factor in the spread of APP was the aerosol spread from infected neighbors.7 In this study, the trading of subclinically infected animals, the frequency of stock purchases, the use of multiple sources, and poor biosecurity were factors associated with the spread of APP in the Danish SPF herds. Most of the herds use AI and bring in pigs in sterile containers. The organism can be spread in the air for a distance of 1 m. Aerosol

transmission of APP9 was possible over 2.5 m. An experiment with transferring air from a group of pigs with serotype 2 showed that if 10% of the air was transferred, then there was no transmission, but if 70% was transferred, then the APP did spread. Experimental aerosol exposure of pigs to serotype 9 results in infection and induces protection to subsequent challenge from the homologous strain. Only a few organisms need to be carried in the tonsil and nasopharynx for a pig to become infectious during travel. Pigs may carry the APP in the nose, and the carriage occurs for both low- and high-virulence strains. The carrier state can be activated by stress or other pathogens. The subclinically infected carrier pig is the most common source of infection. It has been suggested that shedding only takes place at the time of active infection, not when the organism is just carried. Transmission is by the respiratory route, principally via nose-to-nose contact. Overcrowding and inadequate ventilation may facilitate spread. Peak transmission may occur at around 11 weeks. Experimental intranasal challenge has been followed by death in a period as short as 24 hours. The mixing of infected pigs (seeder pigs) with normal susceptible pigs for 48 hours can mimic field infection, with the development of clinical disease, febrile responses, lung lesions, and mortality. The subclinically infected carrier pig is the most common means by which the infection is transmitted between herds. Severe outbreaks may occur unexpectedly in susceptible breeding herds with no previous history of the disease or in intensive feeder pig operations in which pigs are introduced on a regular basis from a variety of sources. Herds that continuously introduce replacement stock are highly susceptible to an outbreak. Following the initial outbreak, general herd immunity develops, but the infection persists, and sporadic cases may continue to occur. The organism is not readily isolated from normal respiratory tissues, but persists in chronic lesions within the lungs of recovered and apparently clinically healthy pigs. These pigs provide a source of infection, especially in a finishing herd buying from diverse sources. The indirect transmission of infection has been proposed but may be rare. An on-farm study described five cases of being transmitted by aerosol or boots or clothes, but the other three cases could have been any combination of these three or even other indirect sources. Risk Factors Pathogen Factors Biotypes There are two biotypes. Biotype 1 requires NAD (NAD dependent) (13 serotypes), and biotype 2 does not (NAD independent; 2 serotypes). Biotype I should be differentiated

from other Actinobacillus species. The isolation of biotype 2 may be increasing. It easily grows on blood agar plates, as does A. suis (see later discussion), which may also under some circumstances produce pleuropneumonia.5 In addition, atypical biotype II strains belonging to APP serotypes 2, 4, 7, 9, and 11 have also been identified.8 In Canada, two biotype I APP13 strains have been found (should be biotype II).9 Serotypes In 1997, two new serotypes were proposed: APP14 and APP15. Serotypes APP1 to APP12 form biotype 1 together with APP15. Biotype 2 is composed of APP13 and 14. Within these categories there are variations because strains may acquire characteristics of other strains. Some of the serotypes are heterogeneous (they share antigenic determinants with other serotypes). Heterogeneity has been reported for APP3, 6, and 8; APP4 and 7; and APP1, 9, and 11. Restriction endonuclease fingerprinting analysis can be used for comparison of serotypes. Serotype 5 is subdivided into subtypes A and B. Serotype 1 has also been divided into antigenic subtypes 1A and 1B. The prevalence of serotypes of APP varies considerably according to geographic location. APP1, 5, and 7 are common in North America; APP2 and 9 are common in continental Europe; and APP3 is common in England and Ireland. APP8 is also found in Ireland. In the British Isles, APP2 and 8 were most common, with 3, 6, and 7 also occurring frequently. APP5, 9, 10, and 12 occurred only rarely. APP1 and 4 were not isolated. In Denmark they routinely find 9 strains of the 15. It is usually APP2 followed by 6, 5, and 12. APP1, 7, 8, and 14 are infrequent, and APP3, 4, 9, 11, 13, and 15 have not yet been found. APP2 is the dominant isolate in Sweden and Switzerland. APP 10 is common in France (also Brazil). APP4 is common in Spain as APP7 and many are nontypeable, but APP4 rarely appears elsewhere.8 And in Spain, biotype II is also quite common. APP1, 7, and 12 are common in Australian pigs, with APP1 being the most common, and APP 15 is also found there and in Japan.10 In North America, the most common serotypes, in order of frequency, are APP1, 5, and 7. APP1 is most common in eastern Canada, accounting for 66% to 83% of the isolates, and is the second most prevalent isolate in western Canada and the United States. APP2 is of low frequency in Canada. However, serotype 2 has now been reported as causing disease in growing and finishing pigs in the United States. Serotype 3 has a low incidence in Canada and the United States. Serotype 5 isolations are common in Canada and the United States. The most common serotypes isolated in Quebec were 1, 5, 2, and 7 in that order. Serotype 6 has not been reported in North America.

Diseases of the Swine Respiratory Tract

VetBooks.ir

The serotyping of isolated strains is important in the epidemiologic and immunologic study of APP infection. It is also important when comparing or analyzing the effectiveness of different treatments to know the virulence of the strains and sensitivity to antimicrobials. An effective immunization program also depends on consideration of the multiplicity of immunogenic types that occur in a particular area or country. It is important to realize that some strains share lipopolysaccharide O-chains and may therefore cross-react. The antigenic crossreactions between APP3, 8, and 15 can also be explained by the presence of structural similarities.11 In one study it was suggested that the presence of APP9 may go clinically unnoticed as M. hyopneumoniae as this potentiates the APP infection.12 Virulence Factors APP attaches to tonsillar epithelium. It also adheres to tracheal rings in vitro and alveolar epithelial cells. Genes that are involved in energy metabolism, nutrient uptake, and stress response are essential for the survival of APP in the pig host. These would include enzymes that are produced in vivo to ensure that there is oxygen. A metalloprotease has been found that can degrade porcine IgA and IgG. Several other virulence attributes and their biological effects have been described. Multiple virulence factors are involved in the development of the disease, and lesions are likely caused by toxic factors associated with the organism. The capsular components are antiphagocytic and inhibit bactericidal activity of the serum but do not cause any lesions themselves. Discovery of mutants without capsules that were no less able to adhere to respiratory tract tissues suggests that the outer membrane proteins were then unmasked (without the capsule), and these were able to adhere to epithelial cells. It is therefore an LPS independent adherence. The outer membrane proteins (OMPs) (60 kDa) adhere to fibers of type III collagen in the lung. The outer membrane proteins appear to be common to all serotypes. Some of the outer membrane proteins are also involved in iron uptake, which is essential for proliferation. The lipopolysaccharides (LPS) of APP are serotype specific but will cross-react with one another. LPS of APP is an important adhesin. It also induces inflammation by stimulating TNF-α, IL-1, Il-6, and IL-8. However, the construction of antibodies to LPS blocked adherence to tracheal cells, so our understanding is not yet complete. The LPS causes an endotoxemia and reproduces certain typical lesions of the natural disease but not the hemolytic or necrotizing effects. Some LPS also help APP to stick to mucus, tracheal rings, and lung, but they do not

1057

Box 12-2  Relative virulence of strains of A. pleuropneumoniae in pigs Very highly virulent

Highly virulent

Moderately virulent

Low virulence

1

2, 4, 6, 8, 15, 9, 11 (10 + 14)

2, 5, 9, 10, 11

3, 7, 12

seem to be involved in adherence to cultured porcine alveolar epithelial cells. Porcine hemoglobin also binds to LPS with APP, and this is a property of the APP OMP. Under iron-deficiency growth conditions, APP expresses 2 transferrin binding proteins. Recently a ferrichrome receptor in APP has been described. There are adhesins (fimbriae) involved in attachment. They are particularly associated with serotype 1 but also 3 and 5 and are usually a feature of subculture 1 (56% of strains), but only 8% on subculture 2 and none on 3. Apx Toxins Not all the differences in virulence are explained by capsules, LPS, hemolysins, and Apx toxins. Certainly, all strains need ApxIV and two out of Apx II or III. There is no certain way to differentiate virulent from avirulent strains Several exotoxins are produced including hemolysins. The hemolytic activity of this organism is characteristic of this species of bacteria. This range of exotoxins is part of the pore-forming RTX group known as the Apx toxins. The latest is Apx IVA, and the gene is present in all APP strains and is species specific and therefore can be used to confirm identification of the organism. The Apx IVA gene is found in all APP serotypes and is absent in the other related species in the Pasteurellacae and, therefore, is considered species specific for APP and is thus being used in a PCR to identify APP strains. It is secreted by a type I secretion system. The Apx toxins are described in the following discussion: I through III can be produced in vitro, but Apx IVa is only produced in vivo and is specific to APP. All 90 strains investigated in one study had Apx IVa genes. Mutants without the capacity to produce Apx toxins do not cause disease. There are basically four different patterns. Both ApxI and II are essential for the production of lesions. Apx III specifically targets leukocytes by binding CD1813 The Apx gene is present in all APP strains. Major RTX toxins in APP are as follows: • ApxI 110 kDa is strongly hemolytic and weakly cytotoxic. • ApxII 102 kDa is weakly hemolytic and moderately cytotoxic. • ApxIII 120 kDa is not hemolytic but strongly cytotoxic. • ApxIV 202 kDa has largely unknown actions but is essential for full virulence of APP.14

Serotypes 1, 5a, 5b, 9, and 11 produce I and II; serotypes 2, 3, 4, 6, and 8 produce II and III; serotypes 7 and 12 produce II; serotype 10 only produces Apx I. There are differences in opinion as to what constitutes virulence. Generally, the following is representative, but it does vary considerably from country to country and isolate to isolate (Box 12-2). These are toxic to alveolar macrophages, neutrophils, and endothelial cells. In small doses they are stimulatory but in large doses lethal. The gene expression is controlled over the growth curve by a novel regulating pathway. Several genes have recently been identified that have helped in survival, including the knowledge that it can produce toxins under anaerobic conditions. The LPS of APP can also stimulate the release of nitric oxide from macrophages by virtue of the enzyme nitric oxide synthase that damages tissues and may disrupt vascular tone, neuronal signaling, and host defense mechanisms. Nitric oxide synthase 2 and cyclooxygenase 2 have been found in swine experimentally infected with APP. Urease activity may also be required for APP to establish infection in the respiratory tract. The increase in antimicrobial drug resistance that has occurred is an indirect virulence factor and an important diseasepromoting mechanism. The ability of the organism to resist complement killing in vitro may reflect a virulence mechanism in vivo that assists bacteria in avoiding the pulmonary defenses of swine and promotes bacterial invasion of the lung. Differences in pathogenicity exist between serovar 1 and serovars 7, 3, and 2. The differences between serotypes 1, 2, and 7 are low. Serotype 3 seems less virulent than 1. The differences in capsular structure and biochemical composition between virulent and avirulent isolates may contribute to virulence. A smooth-type lipopolysaccharide and a rough-type lipopolysaccharide have been isolated and characterized from serotype 5. The intrabronchial infusion of the preparations into pigs induces lesions typical of those in pigs that die of acute pleuropneumonia. APP may interact with P. multocida to produce a severe pneumonia, whereas P. multocida alone is relatively nonpathogenic. Experimentally, a combination of P. multocida and the crude toxin of APP resulted in moderate-to-severe pneumonic pasteurellosis. Of increasing importance and recognition is the formation of biofilms at mucosal

1058

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

surfaces. It is part of the extracytoplasmic stress response to the presence of APP.15 Many strains of APP under appropriate growth conditions form biofilms.16 Serovars 5b and 11 may exhibit biofilm formation, and a histone-like protein H-NS regulates biofilm formation and virulence of APP.17 Animal Risk Factors The major animal risk factors are related to the immune mechanisms and the immune status of pigs of varying ages. A major animal risk factor is that clinically recovered pigs commonly serve as carriers of the organism and never fully recover from the infection. Normally, the APP is detected in mixed bacterial samples from the tonsils and/or nasal samples by PCR from the age of 4 weeks on, but it has been detected as early as 11 days in tonsil samples, so it is possible for the sow to infect the piglet. Isolations become more common from 4 to 12 weeks as maternal antibody wanes. The median length of tonsillar carriage may be 7 to 8 weeks. Colonization of the lungs can develop from around 12 to 16 weeks in some herds to as late as 23 weeks in others. Factors associated with pleurisy in pigs in a case-control analysis of slaughter pigs in England and Wales18 showed that risk factors included the following: • No all-in/all-out policy • Pigs with more than 1-month age difference in the same shed • Repeated mixing • Moving during the rearing phase Decreased incidence was associated with the following: • Grow to finish or wean to finish in a house filled with less than 3 sources • With cleaning and disinfection of grower and finisher groups between groups and extended down time of grower or finisher units Noninfectious factors in the occurrence of pleurisy have been investigated in France.19 This study was in 143 farrow to finish herds, where management, husbandry, and housing conditions were recorded. An increased risk for extensive pleuritis occurred where there was a short temperature range for the ventilation control, lack of disinfection in the farrowing room, late surgical procedures on the piglets, a mean temperature below 23° C (75 F) in the finishing room, and a herd size above 200 sows. Immune Mechanisms Colostral immunity lasts from 2 weeks (usually 5) to 3 months. After an experimental or natural infection antibodies occur 10 to 14 days postinfection and reach their height at 4 to 6 weeks postinfection. In the animals that are subclinically affected there may be no antibodies produced to the toxins. In most herds, high antibody levels in 4-week-old piglets can still be detected, and

this maternal antibody (AB) continues to decrease until about 12 weeks, and then the AB starts to rise with the acquisition of a pathogenic burden. The presence and decay of acquired colostral antibodies between 2 weeks and 2 months determines the age at which APP infection is most likely to occur. The maternal antibody titers halve every 3 weeks and therefore may remain for 12 to 56 days. Nasal colonization can occur as early as 4 weeks, and APP can be found in the lungs from 12 weeks; it is usually 12 to 23 weeks before there is any seroconversion to Apx toxins. In other words, nasal colonization does not always produce antibodies. Active immunity to disease usually follows experimentally induced and naturally occurring infections, and infection with one serotype of APP confers a strong immunity to the same serotype and a partial protection against heterologous strains. Most recovered pigs have a strong humoral immunity but it does not necessarily stop them from becoming carriers and thence possible shedders of APP. Vaccination with killed bacteria produces partial protection against the homologous strain and none against heterologous strains. Second-generation vaccines with Apx toxins produce good protection against clinical disease caused by any serotype but do not prevent animals from becoming carriers through subclinical infections. However, vaccine immunity is serotype specific. The antibody response to APP infections or vaccination is demonstrated by the complement fixation test or other serologic tests. There is a good correlation between a CF titer and resistance to infection, and the organism usually cannot be isolated from seropositive animals. Susceptibility to APP can be predicted by the absence of neutralizing antibodies to the organism, whereas protection can be predicted by the presence of these antibodies. An aerosol exposure of pigs to viable or inactivated serotype 9 induced antibodies in pulmonary fluids and serum, and protected against homologous challenge. However, the organism may persist in necrotic foci in the lungs or tonsils of pigs considered immune to the infection. Within 2 to 3 weeks of an acute disease outbreak, the morbidity decreases because of the development of immunity. Clinical disease is unlikely in adult immune animals, and immune sows confer passive immunity to their piglets that provides protection for the first weeks of life. However, acute disease may occur in piglets 3 to 8 weeks of age if colostral immunity is initially low and wanes to below protective levels. Also, severe cases can occur in nonimmune gilts and boars introduced into infected herds. Pigs infected with hemolytic Actinobacillus spp. may become false-positive reactors for APP. Such pigs may also be less susceptible to pleuropneumonia caused by APP.

Environmental and   Management Factors Outbreaks of the disease appear to occur in pigs that lack immunity, are overcrowded, or have been subjected to recent stressors, such as marked changes in ambient temperature or a failure in the ventilation system. The organism survives better when conditions are wet or in mucus and may last days or even weeks. It survives in water for 30 days at 4° C (39° F) but has a very short survival under dry and warm conditions. Outbreaks may occur in breeding herds following transportation to and from livestock shows and sales. Presumably, the infection was contracted by commingling with clinically healthy but infected pigs. The hypothalamic– pituitary–adrenal axis is stimulated in response to a wide variety of stressors, and this may lead to activation of the organism from the tonsils. The highest risk is associated with the introduction of pigs from sales barns and the lowest risk from stock whose health status is known to the purchaser. Economic Importance The economic losses associated with the disease are considered to be attributable to peracute deaths, the costs of treatment of individually affected pigs and mass medication of the feed and water, and chronic disease that delays the marketing of finishing pigs. Field observations indicate that 5.64 additional days are required for pigs with subclinical infection to reach market weight of 113.6 kg compared with uninfected herdmates. However, other observations and investigations indicate that average daily gain is not significantly affected by infection with APP. Undoubtedly, there are major economic losses associated with the endemic nature of the disease, which is characterized by peracute deaths that recur sporadically, sometimes punctuated by outbreaks.

PATHOGENESIS

The interactions of APP with host epithelial cells seem to involve complex interactions resulting in the regulation of various bacterial genes, including some coding for putative adhesins.20 The natural route of infection is aerogenous. In growing pigs the disease appears to be a respiratory infection without septicemia, producing a fibrinous necrotizing hemorrhagic pleuropneumonia with pleuritis. Early after intranasal inoculation the bacteria were mainly associated with the stratified squamous epithelium and detached epithelial cells in the tonsil. If only a few organisms are inhaled, probably they are trapped in the tonsil and remain there until they are activated. If large numbers are inhaled or if spread from the tonsil reservoir occurs, then a bacteremia probably results. Vacuolation and desquamation of the tonsillar epithelium was observed and there were many migrating neutrophils and these distend the

Diseases of the Swine Respiratory Tract

VetBooks.ir

tonsillar crypts. They do not bind to the tracheal (perhaps in the newborn) or to the bronchial epithelium, but they can stick to the alveolar wall.20 The ApxI of APP10 induces apoptosis in porcine alveolar epithelial cells.21 The adhesion of bacteria to cells appears to be essential and seems to be mediated by polysaccharides and proteins.22 The role of the fimbriae is not clear. Discharge of vesicles containing proteases and Apx toxins from APP1 has been described. Later the bacteria are associated with the crypt walls and detached cells in the crypts. Experimental aerosol exposure of pigs to APP results in a severe fibrinous hemorrhagic necrotizing pleuropneumonia that simulates the natural disease. The organism expresses a number of factors that help to acquire iron and it can use a variety of compounds, including hemoglobin.22 Normally, the APP are kept out of the alveoli by the mucociliary clearance mechanism but not if there are large numbers of APP or there is preexisting damage to the clearance such as occurs as in M. hyopneumoniae infection.12 It is a very determined battle in the alveolus between the APP virulence factors and the host defense mechanisms. The cytokine production excites the defenses and increases the permeability of the alveolar capillary walls and allows access of antibodies and complement. The macrophages need opsonins to help phagocytosis as APP is resistant to the action of complement. The Apx 1 toxin induces apoptosis in the macrophages, which are then killed by leukotoxins, and these then release further amounts of proteases etc. The characteristics of the pathogenesis have been described.23-25 Within a few hours following endobronchial inoculation of various doses of the organism into 12-week-old pigs, clinical evidence of dyspnea and fever are obvious. An aerosol infection with the organism results in pulmonary edema with multifocal petechial hemorrhages and a diffuse neutrophilic bronchiolitis and alveolitis within hours of infection. In the lung, the recruitment of neutrophils is directed toward the viable APP organisms, and possibly 30% of the lung neutrophils respond. This is further enhanced by IL-8 activity. The porcine mononuclear cell phagocytic populations during inflammation produced by APP have been described.26 The lesion is particularly marked in the dorsocaudal regions of the lung. The ability of APP hemolysin to debilitate pulmonary macrophages may enhance the multiplication of the organism, but experimentally the hemolysin of serovar 2 is not an essential factor for the production of the lesions. In the acute stages there are marked vascular changes in the lungs. The lesions resemble infarcts because of the vasculitis, thrombosis, and hemorrhage. There are many necrotic foci that serve as reservoirs of the organism in pigs that recover. In the experimental disease, the leukogram is typical of acute inflammation; however, hypoxemia and

alveolar hypoventilation are not features of the disease. The hematologic and physiologic findings indicate that the peracute disease resembles septic shock. Immediately after infection the levels of IL-1, IL-6, and TNF-α begin to rise. Moderate levels help in defense, but high levels make things worse. At the same time the IL-10 suppresses TNF-α and IL-1 production in macrophages and monocytes, which up-regulates the other inflammatory cytokines. Pretreatment of the pig with IL-10 reduces the severity of the pleuropneumonia. The prolonged survival of APP during the infections may be attributable to the effect the organism has in downgrading the protective responses of the host. The distribution of porcine monocytes in different lymphoid tissues and the lungs during experimental. A. pleuropneumoniae infection and the role of chemokines has been described.27 This study showed that monocyte counts in various organs changed during inflammation. The CD163 + monocyte counts were found in the lungs and TBLN from APP-infected pigs, suggesting that monocytes migrate just to these organs.

CLINICAL FINDINGS

The clinical signs vary with the immune status and environmental stress and customarily may be seen between 6 and 20 weeks of age. In all cases there is a reduced growth rate and reduced feed intake, therefore leading to reduced weight gain. There is no relationship between average daily gain and serologic response to APP. The illness may be peracute, acute, subacute, or chronic. In all stages there is very little exercise tolerance, with varying degrees of increase in respiratory rate. The onset is sudden. Several pigs that were not seen ill may be found dead, and others show severe respiratory distress. Affected pigs are disinclined to move and are anorexic. A fever of up to 41° C (105.8° F) is common, and labored respirations with an exaggerated abdominal component (‘thumps’), cyanosis, and frequently a blood-stained frothy discharge from the nose and mouth are characteristic, particularly just before death. In peracute cases, the clinical course may be as short as a few hours, but in the majority of pigs it is 1 to 2 days. In many cases, the animals “dogsit” with elbows abducted to relieve pressure on the lungs, and they show dyspnea. Chronic cases, which usually appear after the acute phase has disappeared, are febrile and anorexic initially, but respiratory distress is less severe, and a persistent cough may develop. If affected pigs are not treated, there will be a high case-fatality rate. Otitis media in a weaned pig caused by infection of the middle ear with the organism has been described. There may also be lesions in the joints with fluctuating swellings of the hocks and the synovial membranes replaced by granulation tissue.

1059

The course of the disease in a herd may last for several weeks, during which time new acute cases develop and chronic cases become obvious by an unthrifty appearance and chronic coughing. Abortions may occur and the disease may cause sudden deaths in adult pigs, particularly those that are kept outdoors during the summer months and exposed to very warm weather. Computer tomography and radiography have been described as aids to diagnosis.28 Recently a very mild condition very similar to swine influenza, with just a slight increase in respiratory rate, has been described.29

CLINICAL PATHOLOGY

Plasma cortisol rises 24 hours postchallenge. Haptoglobin is increased. Within 48 hours IL-1a, IL-1β, and IL-8 were increased, and there was a 50% reduction in iron and zinc. Plasma IGF-1 concentrations were reduced in response to the APP challenge as they were with endotoxin challenge. The LPS of APP produces rises in inflammatory cytokines (TNF-α, IL-6, and IL-10). Band neutrophils are significantly increased in early infections from 18 to 48 hours, and the early changes have been described.30 Culture of Organism In an outbreak, the diagnosis is preferably made by culture at necropsy. Carrier pigs can be identified by culturing the organism from the upper respiratory tract using nasal swabs from live pigs on the farm and samples from tonsils at slaughter. A selective medium for the culture of the organism from the airways of slaughtered pigs may increase the isolation rate because of the high degree of contamination. The culture of APP has recently been complicated by the identification of the nonpathogenic A. porcitonsillarum. Serotype of Organism Tests to determine the serotype include slide agglutination, immunodiffusion, ring precipitation, indirect hemagglutination, immunofluorescence, coagglutination, and counterimmunoelectrophoresis. The latter is quicker, more sensitive, and more easily performed than direct immunofluorescence and immunodiffusion procedures. The coagglutination test is simple and rapid, the immunodiffusion test is considered to be the most serotype-specific, and there is a good correlation between the rapid slide agglutination test and the indirect fluorescent antibody tests. The rapid slide agglutination test is the method of choice of some workers, but the coagglutination test is serotype-specific, sensitive, simple, rapid, reproducible, and easier to read and interpret than the rapid slide or tube agglutination tests. The International Pig Veterinary Society has recommended that the coagglutination test is currently the method of choice for routine

1060

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

serotyping of field strains. This technique does not allow separation of the heterogeneous serovar 8 from serovars 3 and 6, the heterogeneous serovar 9 from serovar 1, or the heterogeneous serovar 7 from 4. The results are reported as group 9–1, group 8–3–6, and group 7–4, respectively. The final identification of heterogeneous serovars can only be achieved by the agar gel diffusion test and by indirect hemagglutination. Reference strains and the corresponding antisera are available to bring some uniformity into serotyping. Detection of Antigen The polymerase chain reaction (PCR) is a highly sensitive test for the detection of the organism from tissue samples. A PCR for type 4 has been developed. Some detect OMP; others detect Apx genes. Apx IVA based ELISAS can be used for evaluating APP status in commercial herds, but some appear limited by high carriage rates of lowvirulence APP.31 Immunomagnetic separation of APP1 and 2 has been described with greater sensitivity than possible with isolation or even PCR. A PCR-based RFLP analysis of the OM1A gene may also be of value in differentiating APP serotypes. A multiplex PCR has been developed. There is often disparity between immunologic and PCR-based serotyping.32 Serology Serology is the best method for surveillance purposes and is the best way to detect subclinical infections but may give unexplained results.33 In addition, some of the strains do not produce ApxIV do not produce antibodies.34 Sometimes diagnostic interpretation is difficult.22,33 Tests for antibodies to toxins and/or capsular antigens have a low specificity and can also be positive for A. suis infection. Most commonly used are antigens using O-chain LPS.35 They tend to be grouped together: (1, 9, 11), (2, 3, 6), (8, 4, 7), (10 and 12), (3 and 5), (15, 3, 6).11 For the serologic diagnosis of infection in live animals the complement fixation test is reliable, but an enzyme-linked immunosorbent assay (ELISA) test is highly specific and more sensitive than the complement fixation test. The complement fixation test has been used routinely in the past in some countries and has a high degree of sensitivity and specificity. It is, however, a cumbersome test, and many laboratories find it difficult to perform, and so it is rarely used nowadays. Pigs being imported into China and Russia still require a CFT negative test. The ELISA is a rapid and sensitive test and can be adapted to automation. The ELISA for serotypes 1, 2, 5, and 7 distinguishes exposed from unexposed pigs or herds. Because of cross-reactivity with other serotypes and A. suis, the serodiagnosis of

serotype infections cannot be made with certainty. A blocking ELISA is available for detection of antibodies against serotype 2 and also 2, 6, 8, and 12, which is the dominating serotype in Danish swine herds, causing approximately 70% of diagnosed outbreaks of pleuropneumonia. A similar test is available for serotype 8. A mixed-antigen ELISA for serodiagnosis of serotypes 1, 5, and 7 has a sensitivity of 96% and specificity of 99.5% and can be used for herd health monitoring programs. The long-chain lipopolysaccharide of serotypes 4, 5, and 7 is a superior antigen to the crude extracts used as antigens in the ELISA for the serodiagnosis of pleuropneumonia. There are now ELISAs for the detection of antibodies to the Apx toxins, and the one for type II Apx was described as sensitive, inexpensive, and highly discriminatory. A multiplex PCR for all toxins in one test is a reliable typing system. A new ELISA for the Apx IV produced by all 15 serotypes means that you can detect all APP with one test. It has a specificity of 100% and a higher sensitivity than culture (93.8%). This is important because you can find Apx I to III in pigs associated with A. suis and A. rossii, but Apx IV is only produced by APP in vivo. It will detect the toxin from 2 to 3 weeks postinfection. An inhibition enzyme immunoassay for the serodiagnosis of serotypes 2 and 5 had a sensitivity and specificity of 100% and 98.9%, respectively. The detection of antibodies to APP is an essential feature in the epidemiologic study and control of pleuropneumonia in pigs. Serologic testing can be used to monitor the level of infection in a breeding herd over a period of time and as the piglets become older. A minimum of 30 serum samples from adult pigs is necessary to provide a reliable assessment of the herd’s infection status. None of these serologic tests is completely reliable, and in certain situations a combination of two tests is needed for interpretation of low titers in some pigs. In most instances, serologic diagnosis is typespecific, and protection obtained by vaccination is type-specific and will protect only against the serotype contained in the vaccine. Thus it is important to determine the serotypes that are causing disease in the herd. An important strategy of control of this disease is to detect infected pigs in a herd or to exclude infected pigs from being imported into a herd. Because there is no reliable method for the detection of every infected pig, the effectiveness of this barrier is reduced whenever pigs, such as breeding stock or weanlings, are allowed into a herd. There is a need for a highly sensitive and specific test for the identification of infected pigs. Although bacteriologic culture is specific it is not sensitive. The ELISA test may be a useful test for the antemortem diagnosis of infected herds.

NECROPSY FINDINGS Characteristic lesions are confined to the thoracic cavity and consist of hemorrhagic and fibrinous pleuropneumonia with a tendency to sequestration in the chronic form. In peracute cases the lungs are swollen, firm, and dark red. In peracute cases the trachea and bronchi are full of frothy fluid. Fluid and blood ooze from the cut surface, and there may be marked edema of the interlobular septa, reflecting widespread thrombosis and alterations in capillary permeability. There may be hemorrhagic areas of necrosis that are very variable. In acute cases there are layers of fibrin on the pleural surface and pericardium. In pigs that die less acutely, focal black or red raised areas of pneumonia are present. Lesions may occur throughout the lung, including the diaphragmatic lobes. The quantitative morphology of peracute pulmonary lesions induced by the organism has been described. In chronic cases there is fibrosis of the fibrinous pleurisy and adhesions result between the visceral and parietal pleura, and on removal of the lungs from the thorax portions of lung may remain adherent to the thoracic cage.36 A fibrinous pleuritis overlies the affected lung tissue, and a fibrinous pericarditis may also be present. The organism can be isolated from affected lung tissue, but generally not from other internal organs. Occasionally, otitis, endocarditis, pericarditis, and serous arthritis may follow, particularly when infection involves serotype 3. An osteomyelitis and arthritis caused by APP has been demonstrated using fluorescent in situ hybridization. Histologically, vasculitis and widespread thrombosis is usually evident, in addition to an abundance of fibrin and neutrophils within alveoli. A fibrinous thrombosis with IHC demonstration of APP has been described. In situ hybridization can be used to detect IL-1, IL-6, and TNF-α in streaming degenerate alveolar leukocytes (oat cells) and the boundary zone of oxidative necrosis. A less intense signal was seen in the dense zone of degenerate cells in granulation tissue surrounding the necrotic areas. IL-1 was also seen in the scattered endothelial cells bordering zones of coagulative necrosis. IL-6 is the cytokine that is most elevated, and serum amyloid and haptoglobin are also elevated. In a chronically infected herd, fibrous pleural adhesions may be present in a large proportion of the pigs at market as a result of infection several months earlier. Subacute to chronic lung lesions are encapsulated by fibrous tissue, and sequestra may be present. A high prevalence of fibrous or fibrinous pleuritic lesions on inspection at the abattoir is very suggestive of APP infection.37,38

DIAGNOSIS

The provisional diagnosis of pleuropneumonia associated with APP in the pig is usually based on history, clinical signs, and the postmortem picture. The acute cases then require

Diseases of the Swine Respiratory Tract

VetBooks.ir

laboratory investigation to confirm, and chronic cases may prove antigen negative (the lesions are usually fibrous or fibrinous) but possibly antibody positive. A variety of samples need to be taken from acute cases and should be from lesions not from inflammatory exudates and particularly not from the lungs. Samples for Confirmation of Diagnosis The evolution of diagnostic tests has been described as follows:39 • Bacteriology—lung culture is relatively easy if the carcass is freshly dead. The culture is achieved on 55 sheep blood agar with a cross-streak of Staphylococcus epidermidis or S. aureus. The plates are incubated overnight with 5% CO2, and a clear zone of complete hemolysis results. Typing will confirm the identity of APP1-15 and if atypical PCRs can be used. PCRs for 3, 6, and 8 were described;40 1, 7, and 12;41 15 and 7;42 and also 1, 2, and 8. Sometimes serotypes cannot be differentiated. Toxin typing using a PCR can be used to determine which Apx toxin genes are carried by a certain isolate. They can also be isolated from pure and mixed bacterial cultures by immunomagnetic separation. • Histology—formalin-fixed lung (LM). APP can be further identified by IHC, which is particularly useful in chronic cases and ISH. • Serology—used to check the herd status. Coagglutination can be used first, with confirmation by agar gel diffusion and indirect hemagglutination.

DIFFERENTIAL DIAGNOSIS The rapidity of onset and spread with fever, anorexia, severe dyspnea, and high mortality differentiates APP from the majority of respiratory diseases in pigs. Enzootic pneumonia is more insidious in its occurrence and has distinctively different epidemiologic, clinical, and pathologic features. Pasteurellosis is characterized by a necrotizing bronchopneumonia. Swine influenza is characterized by an explosive outbreak of respiratory disease. However, this is not restricted to growing pigs and the mortality is low. There is a distinct difference in the respiratory lesion on necropsy examination. Glasser’s disease is characterized by serositis, arthritis, and meningitis, and occurs in younger pigs. Mulberry heart disease may present with similar clinical findings, but there is no pneumonia on necropsy examination.

A. porcitonsillarum also produces and secretes ApxII by an operon that does not occur in APP Actinobacillus suis shares cross reactions with APP 3, 6, and 8. Actinobacillus lignieresii have some cross reactions with APP serotypes.

TREATMENT Antimicrobial Therapy The results of treatment are often disappointing because of the severity of acute disease and persistence of infection in recovered pigs. It is best to assume that APP cannot be eliminated using antibiotic therapy.41 Although antimicrobials may reduce mortality and improve average daily gain, treated animals often continue to harbor the organism and are a source of infection to other animals. If animals are clinically ill, then injection of antimicrobials is necessary. Affected and in-contact pigs should be treated parenterally with antimicrobials. Tetracycline, spectinomycin, and penicillin have been effective and are recommended unless drug resistance has occurred. Penicillin may have inconsistent results.43 Fluoroquinolones are distributed to bronchial secretions, bronchial mucosa, and alveolar macrophages. The pharmacokinetics of danofloxacin are favorable for APP treatment. In fact, elevated C-reactive protein, IL-6, and haptoglobin (all elevated rapidly after infection) all return to normal, as do the reduced plasma zinc, ascorbic acid, and alpha tocopherol rapidly after treatment. Ceftiofur and fluoroquinolones were the most active agents against APP. APP is only eliminated from the respiratory tract in animals medicated with enrofloxacin. Tilmicosin is useful for treating outbreaks. In a large study in Switzerland of 83APP and 58 A. porcitonsillarum (PT) strains screened for susceptibility to 20 antimicrobial agents, it was found that there was resistance to sulphamethoxazole, sulphonamide–trimethoprim, tiamulin, tilmicosin, tetracycline, and ampicillin. A few of the PT strains showed increased susceptibility to enrofloxacin.44 Both APP and PT remain susceptible to cephalosporins, fluoroquinolones, and phenicols, which are not be used except in special cases. In the last few years resistance to tetracyclines and trimethoprimsulphonamide has increased.45-47 There is no clear association between antimicrobial susceptibility and serotype.44 There have been enrofloxacillin-resistant APP isolates found in Taiwan.48 In finishing units, where outbreaks of the disease have been confirmed, the twice-daily intramuscular injection of pigs early in the course of the disease with antimicrobials, based on drug sensitivity tests, daily until clinical recovery occurred, was superior to the mass medication of feed and water. A

1061

considerable amount of labor is required, but it is considered to be the most cost-effective method. In a study of SPF pigs experimentally infected with APP2 and treated with enrofloxacin(E), tetracycline(T), or penicillin(P) at the onset of disease or left untreated, it was found that the animals treated with E and T recovered rapidly. All except the E group developed antibodies. They were later challenged with APP2 again, but here the E group developed serious disease. The implication is that the E was so successful initially in eradicating the APP that it did not allow an antibody response to develop to resist the rechallenge.43 Antimicrobial Sensitivities The antimicrobial sensitivities of isolates of APP have been monitored, and there is some variation based on geographic location. The large expansion in the size of swine herds, and the introduction of breeding stock from many different sources, has led to an increase in the incidence of porcine pleuropneumonia and extensive use of parenteral antimicrobials. To ensure an optimal response to therapy, it is necessary to monitor antimicrobial sensitivity on a herd basis. The antimicrobial sensitivity of the organism was determined in isolates from Europe, Japan, South Africa, and North America between 1989 and 1991. They were highly susceptible to danofloxacin and moderately susceptible to amoxicillin, ceftiofur, and trimethoprim–sulfamethoxazole. There was widespread resistance to other currently available antimicrobials. In another study, thiamphenicol and metronidazole had good activity, and the cephalosporins and fluoroquinolones were most active. A comparison of the minimum inhibitory concentrations (MICs) of several antimicrobials against several bacterial pathogens of swine, including APP, from the United States, Canada, and Denmark found that ceftiofur and enrofloxacin were the most active antimicrobials. Plasmid-mediated antimicrobial resistance has been found in isolates of the organism that are resistant to certain antimicrobials. Antimicrobials in Experimental Disease In these experimental infections enrofloxacin and ceftiofur are particularly effective and also tulathromycin.49 The therapeutic efficiency of some commonly used antimicrobials has been evaluated for the treatment of experimentally induced pleuropneumonia using serotype 1 APP. Florfenicol in the feed at 50 ppm prevented pneumonia when pigs were experimentally inoculated with serotype 1, 2, and 5 strains and thiamphenicol-resistant strains of the organism. The combination of trimethoprim and sulfamethoxazole is superior to

1062

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

a combination of trimethoprim and sulfadimethoxine. Oxytetracycline in the water at 222 mg/L for 7 days beginning 24 hours before experimental challenge reduced the case-fatality rate, lung lesions, and the isolation of the organism compared with the unmedicated group. Treatment of chronically affected pigs did not improve rate or gain, nor did it eliminate the infection. Enrofloxacin at 150 ppm in the feed provided effective control of the experimental disease. Mass Medication of Feed In-feed medication with sulfadimethoxine and sulfamethoxazole in combination with trimethoprim has been described. Oxytetracycline in the feed at 1600 mg/ kg of feed for 6 days before experimental challenge and for 9 days after challenge, provided 100% protection from clinical disease, but 400, 800, or 1200 mg/kg of feed did not prevent subsequent shedding and transmission to seronegative animals. Tetracycline should be administered through the feed of all in-contact pigs during the outbreak, but the persistence of the organism in chronically affected pigs may result in clinical disease when the medication is withdrawn. Doxycycline in feed at 250 ppm for 8 consecutive days is useful for the control of APP. Tilmicosin fed to pigs at 200 to 400 µg/g is effective in controlling and preventing APP-induced pneumonia, using seeder pigs, when administered in the feed for 21 days. In commercial herds, 400 µg/g of feed for 21 days is no more effective than 200 µg/g of feed for the control of naturally acquired pneumonia caused by APP and P. multocida. Sulfathiazole at the rate of 28 g/3.8 L of drinking water for 12 days has also been successful. Tiamulin in the drinking water at a concentration to deliver 23 mg/kg BW for 5 days after an initial individual treatment of affected pigs has also been recommended.

CONTROL

It is impossible to guarantee freedom because the detection of carriers is almost totally impossible. There are no techniques as yet for identifying the animal that may have only a few organisms in the tonsil. You can guarantee freedom from clinical disease at the time of inspection but little else. In a recent study of 980 pigs there was no evidence of an APP clinical or pathologic case until the occurrence of PMWS resulted in the isolation of an APP7 from the series of pigs in a unit that had until that time been considered free. There are two options for the control and prevention of porcine pleuropneumonia: 1. Control at an economical level using good management combined with the possible use of vaccines 2. Eradication of the infection from the herd

Determining which option to select requires careful consideration of the advantages and disadvantages of each option. With an understanding of the factors that result in clinical disease, it is possible to maintain an infected breeding herd and produce pigs with a small risk of clinical disease. Control by Management Management and housing improvements can prevent clinical episodes. One of the most important things to do is to make sure that there is vaccination for enzootic pneumonia. Control is difficult and unreliable because pigs that recover from clinical disease provide a source of future infection for finishing operations that purchase all of their introductions. The all-in, all-out system of purchasing, feeding, and marketing pigs, with a thorough cleaning between groups of animals in a finishing operation, should be adopted. The disease is highly contagious, and control measures must be directed toward identifying infected pigs and eliminating their introduction into uninfected herds. When moving pigs between herds, it is critically important that the herds be matched according to their infection status. Source herds for feeder animals are serotested, and then pigs of like immune status are commingled to produce a population that is compatible immunologically. By commingling only animals from seronegative herds, the risk of disease is greatly decreased and growth performance improved. The mixing of animals from herds known to be infected with homologous serotypes is also effective. Every economical effort must be made to identify and isolate infected pigs and to exclude the importation of clinically normal but infected pigs into herds in which the infection is not present. This is a major challenge that is dependent on the availability of a highly sensitive and specific laboratory test. The acquisition of new breeding stock for herds free of the infection should include a period of quarantine and two serologic tests 3 weeks apart. Only seronegative animals should be introduced into the herds. A seropositive animal should be considered a potential carrier. Field trials have shown that it is not possible to rear seronegative animals within a breeding and rearing herd heavily infected with serotype 2 of the organism. Neither medication of the sows and piglets with trimethoprim–sulfonamides, nor a strictly applied all-in, all-out system reduced the percentage of seropositive animals. Management practices must emphasize the rearing of weaned pigs in pens separate from older stock that are carriers of the organism. Large breeding herds and finishing units should subdivide the total herd into separate units, which minimizes the spread of infection. Early weaning and segregation

of gilts from infected stock have been used to develop a seronegative herd. Herds can be classified into one of three categories depending on their infection status: CATEGORY 1. Serologically positive for APP without a history of clinical disease. A majority of herds are serologically positive but do not have clinically apparent disease. Good management and environmental quality control can minimize the incidence of clinical disease. Good ventilation, the use of all-in, all-out management practices, and appropriate stocking densities are important. CATEGORY 2. Serologically negative and clinically free of APP. These herds can be maintained free of infection with good biosecurity practices. New breeding stock must be obtained from herds free of infection. Artificial insemination can be used to limit the introduction of live pigs. Pigs sold from these herds to herds with endemic infection are highly susceptible to infection. CATEGORY 3. History of clinical disease caused by APP, which has been pathologically and microbiologically confirmed. In these herds, acute disease outbreaks occur most commonly in pigs 9 to 20 weeks of age. Pigs are usually protected by colostral immunity for the first 8 weeks of life. The severity of outbreaks can be reduced by mass medication of the feed, treatment of individual pigs, and good management practices to ensure adequate ventilation. Eradication The Danish SPF system was the first to try to eradicate APP. Each month 20 serum samples were tested for APP 2 and 6 and were collected at the monthly clinical inspection. This happened every 3 months also for APP12 and annually for APP 1, 5, 7, and 10. Recurring outbreaks of pleuropneumonia is the most common reason for an eradication strategy. Eradication is done by depopulating the entire herd, followed by repopulation with animals from herds that are clinically and serologically negative. Eradication can be successful but the risk of introducing infections into the herd is high unless biosecurity measures are adopted and strictly implemented. An alternative to depopulation is medicated early weaning, in which pigs are weaned at 10 to 15 days of age, treated with antimicrobials, and reared in isolation. Transmission of infection between the sow and piglets does not occur before 11 days of age, about half of the piglets are infected at 16 days of age, and if weaned at 21 days of age most of the piglets are infected.

Diseases of the Swine Respiratory Tract

VetBooks.ir

The early weaning program can be expanded to the three-site system of rearing. Adults and nursing piglets are housed in one site. At weaning, piglets are moved to the nursery barn for growth to 25 kg, and then moved to a third site for the final growing period. The adults may be serologically positive for infection, but the nursery pigs, growing pigs, and finishing pigs are negative. Age segregation, distance that prevents aerosol transmission, and adherence to strict biosecurity practices can reduce the prevalence of infection and the incidence of disease. Vaccination A wide range of vaccines have been developed over the years.50 There are two main groups of vaccines. One is killed organisms the bacterins, and these are serotype specific. The second group are subunit toxin-based vaccines. These contain Apx I, II, III with or without OMP and show a high degree of protection all APP1-12 serotypes. The in vivo ApxIV works well but has not yet been commercialized,48 although ApxIV is not needed for effective vaccination. An Apx 1A mutant has potential for a live attenuated APP vaccine.51 Animals vaccinated with bacterins will produce antibodies that will cross-react with ELISA tests that use polysaccharides as antigens. There is a considerable effect of adjuvants in these vaccines. Natural or experimental infection with a serotype of APP induces a strong immunity to both homologous and heterologous serotypes. Vaccination has been attempted to prevent pleuropneumonia in pigs. However, the protection obtained by parenteral vaccination is serotype specific, and vaccines must therefore contain the serotype existing in the swine population. The mortality rate is lower in vaccinated animals, but they are still carriers of the organism. Serotype 8 is closely related to serotypes 3 and 6, and parenteral revaccination using a capsular extract or killed APP serotype 8 provides a high degree of protection against challenge with serotypes 3 or 6. A tetravalent vaccine containing serotypes 1, 2, 5, and 7 stimulated titers to all four serotypes and an anamnestic response was induced by a second vaccination. This suggests that the serologic and cross-protective properties of APP serotypes should be identified before they are used as antigen in the complement fixation test and in vaccines. The protein associated with the capsule of APP is responsible for serotype-specific protection against mortality in mice. Further purification and characterization of this protein antigen is needed to determine whether it is the specific antigen responsible for protection against mortality in swine or if it is a necessary carrier for a serotype-specific capsular disaccharide antigen. The vaccines that have been evaluated are killed vaccines with an adjuvant. In one

experimental trial, two and three vaccinations using a bacterin containing serotypes 1 and 5 prevented mortality following an aerosol challenge with the same serotypes as present in the vaccine. However, all vaccinated pigs had severe signs of respiratory disease and the vaccine did not prevent the development of lung lesions. The use of a formalin-inactivated alum-precipitated vaccine containing serotype 1 was effective in decreasing the morbidity and mortality rates from naturally occurring pleuropneumonia. The adjuvanted vaccines have caused considerable tissue reaction, resulting in abscesses and granulomas. The mineral oil adjuvants are highly irritant and cause granulomas, which are present 8 weeks after vaccination but result in high titers. The aluminum hydroxide adjuvants are less irritating but result in lower titers. Vaccines containing a lecithin-base oil at 5% are nonirritating and stimulate high complement fixation titers. Subunit vaccines containing purified or partially purified antigens provide better protection than whole cell vaccines. Capsular antigens, outer membrane proteins, lipopolysaccharide, and soluble toxic factors are immunogenic in pigs. An acellular vaccine containing multiple virulence factors provided complete protection from mortality and significantly reduced morbidity to homologous challenge. Pigs vaccinated with the cell extract had fewer clinical signs of pleuropneumonia than pigs vaccinated with three other commercial vaccines and challenged with serotype 1. A vaccine containing the LiCi cell extracts and a crude hemolysin isolated from serotype 1 provided protection against both mortality and morbidity in vaccinated pigs challenged by intratracheal inoculation. An experimental vaccine using bacterial “ghosts,” which are empty cells produced by bacteriophage lysis appears to be successful. A better cellular response was observed to inactivated bacteria than to ghost vaccines. Bacteria grown in conditions resulting in high in vitro adhesin levels induced better protection than those grown in NAD rich medium. An APP type 2 vaccine has been described with deletions in the Apx IIA gene, which can then function as a negative marker vaccine, which appears to be capable of protecting pigs without shedding. Antigenic variation within a capsular serotype, for example in subtypes 1A and 1B, as a result of antigenic variation within the lipopolysaccharide, can result in the failure of whole cell bacterins to provide protection against the same capsular serotype. This lack of cross-protection within a capsular serotype provides a partial explanation for vaccination failures observed under field conditions. A polyvalent bacterin containing serotypes 1, 3, 5, and 9 provided satisfactory protection against homologous challenge 14

1063

days after the second vaccination. Mortality was reduced, and lung lesions, pleural adhesions, and isolations of the organism from the tonsils and lungs were reduced. It is possible in the future that a differentiation from vaccinated animals test may be based on the ApxIVA gene.52 Live vaccines using laboratory-obtained nonvirulent mutants have also been developed and shown to protect against homologous and heterologous serotypes.53-55 REFERENCES

1. Vengust G, et al. J Vet Med B Infect Dis Vet Publ Hlth. 2006;53:24. 2. Hoeltig D, et al. BMC Vet Res. 2009;5:14. 3. Sjolund M, Wallgren P, et al. Acta Vet Scand. 2010;52:23. 4. Opriessnig T, et al. Anim Hlth Res Rev. 2011;12:133. 5. MacInnes JI, et al. Can J Vet Res. 2008;72:242. 6. Kokotovic K, Angen O. J Clin Microbiol. 2007;45:3921. 7. Zhuang Q, et al. Vet Rec. 2007;160:258. 8. Maldonado J, et al. J Vet Diag Invest. 2009;21:854. 9. Gottschalk M, et al. Proc Cong Int Pig Vet Soc. 2010a;21:290. 10. Koyama T, et al. J Vet Med Sci. 2007;69:961. 11. Gottschalk M, et al. Proc Cong Int Pig Vet Soc. 2010b;21:289. 12. Marois C, et al. Vet Microbiol. 2009;135:283. 13. Vanden Bergh PG, et al. Vet Res. 2009;40:33. 14. Liu JL, et al. Vet Microbiol. 2009;137:282. 15. Bosse J, et al. J Bacteriol. 2010;192:244. 16. Labrie J, et al. Vet Res. 2009;41:03. 17. Dalai B, et al. Microb Pathogen. 2008;46:128. 18. Jager HC, et al. PLoS ONE. 2012;7:e29655. 19. Fablet C, et al. Epidemiol Sante Anim. 2013;63:13. 20. Auger E, et al. Infect Immun. 2009;77:1426. 21. Chien M-S, et al. Vet Microbiol. 2009;135:327. 22. Chiers K, et al. Vet Res. 2010;41:65. 23. Foote SJ, et al. J Bacteriol. 2008;190:495. 24. Goure J, et al. BMC Genomics. 2009;10:88. 25. u Z, et al. PLoS ONE. 2009;3:e1450. 26. Ondrackova P, et al. Vet Res. 2010;41:64. 27. Ondrackova P, et al. Vet Res. 2013;44:98. 28. Brauer C, et al. BMC Vet Res. 2012;8:47. 29. Tobias TJ, et al. Vet Rec. 2009;164:402. 30. Hedegaard J, et al. Acta Vet Scand. 2007;49:11. 31. Eamens GJ, et al. Aust Vet J. 2012;90:225. 32. O’Neill C, et al. Vet Rec. 2010;167:661. 33. Broes A, Gottschalk M. Proc Ann Meet Am Assoc Swine Vet. 2007;193. 34. Tegetmeyer HE, et al. Vet Microbiol. 2009;137:392. 35. Klausen J, et al. J Vet Diag Invest. 2007;19:244. 36. Merialdi G, et al. Vet J. 2012;193:234. 37. Fraile L, et al. Vet J. 2010;184:325. 38. Meyns T, et al. Vet J. 2011;187:388. 39. Costa G, et al. Vet Microbiol. 2011;148:246. 40. Zhou L, et al. J Clin Microbiol. 2008;46:800. 41. Angen O, et al. Vet Microbiol. 2008;132:312. 42. Ito H. J Vet Med Sci. 2010;72:653. 43. Sjolund M, et al. Vet Rec. 2009;164:550. 44. Matter D, et al. Vet Microbiol. 2007;122:146. 45. Gutierrez-Martin CB, et al. Vet Microbiol. 2006;115:218. 46. Hendricksen RS, et al. Acta Vet Scand. 2008;50:19. 47. Marioka A, et al. J Vet Med Sci. 2008;70:1261. 48. Wang Y-C, et al. Vet Microbiol. 2010;142:309. 49. Hart F, et al. Vet Rec. 2006;158:433. 50. Ramjeet M, et al. Anim Hlth Res Rev. 2008;280:39104. 51. Xu F, et al. Vet Microbiol. 2006;118:230. 52. O’Neill C, et al. Vaccine. 2010;28:4871. 53. Bei W, et al. Vet Microbiol. 2007;125:120.

1064

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

54. Lin L, et al. FEMS Microbiol Lett. 2007;274:55. 55. Park C, et al. J Vet Med Sci. 2009;71:1317.

MYCOPLASMA PNEUMONIA (MYCOPLASMA HYOPNEUMONIAE) ETIOLOGY

Mycoplasma hyopneumoniae (once also called Mycoplasma suipneumoniae) is the primary causative agent. M. hyopneumoniae (MH) inhabits the respiratory tract of pigs, appears to be host specific and survives in the environment for only a very short period of time. The disease has been reproduced with pure cultures, and the organism can be demonstrated directly or indirectly in pigs with enzootic pneumonia worldwide. The isolation of MH is complicated by the presence of other mycoplasmas in the upper respiratory tract of pigs including M. hyopharyngis, M. hyorhinis, M. sualvi, and Acholeplasma species. The nonpathogenic M. flocculare also complicates the culture of M. hypneumoniae. The strains of MH are antigenically and genetically diverse. Multilocus sequence typing has been used to estimate genetic diversity,1,2 and it showed that specific MH strains are responsible for local outbreaks as they are in geographic contact or operative contact. A wide variety of genetic diversity was found in U.S. strains using comparative genomic hybridization. Significant variation at the genetic level has also been found;3 and it has not yet been established as to what constitutes cross protection and virulence.4 MH varies its surface proteins through varied proteolytic events.5,6 A proteonomic survey of MH identified a total of 31 different coding DNA sequences.7 Genotyping of MH in wild boar samples showed that variability was high, but there was geographic relatedness; they were related to the domestic pigs, but no matching types were found.8

EPIDEMIOLOGY Occurrence and Prevalence of Infection Enzootic pneumonia occurs in pigs worldwide, and the incidence is high in intensive pig rearing enterprises. Lesions may be present in 40% to 80% of the lungs of pigs at abattoirs. The peak incidence of pneumonia occurs at 16 to 20 weeks of age, which is likely related to the increased stocking density in this period. In northern climates, the incidence of clinical disease and prevalence of lesions at slaughter are higher in the summer months. The prevalence of lung lesions is often highest in pigs slaughtered in the winter months compared with autumnslaughtered pigs. The amount of bronchopneumonia lesions in individual lungs ranged from 0% to 69%, with an average of 7.8%. A 2002 survey in the United States showed that 82.3% of finishing sites had at

least one animal positive on antibody testing and 94.4% of breeding sites. Seroprevalences were higher in the clinically affected herds, and most of the pigs were infected with MH at a younger age. A study in the United Kingdom showed that geographic location of the finishing unit appeared to be a statistically significant risk factor for EP-like lesions and pleurisy.9 In addition, they also found that part-slatted floors were a potential risk factor. In a study of colonization at weaning and the infection at slaughter, average lung lesion scores, percentage of affected lungs, presence of MH on the bronchial epithelium, and seroconversion, it was found that the severity of the disease can be predicted by the prevalence at weaning in segregated systems. Strategies focused on reducing colonization at weaning may help to control MH in segregated production systems.10 Vaccination does not prevent transmission to sentinel pigs in contact with infected animals. Transmission of MH from asymptomatic carriers to unvaccinated and vaccinated sentinels was not different.11 Morbidity and Case Fatality In infected herds, the morbidity rate is high during the growing period, but the casefatality rate is low. There is however, an increase in the number of treatments of sick pigs in comparison with herds free from the disease. The morbidity rate falls markedly with increasing age, and there is a much lower incidence of pneumonic lesions in sows, even though they may still harbor the organism. However, when MH gains entry into a herd that has been previously free of the disease, all ages of pigs are affected, and mortality, even in adults, can occur. Methods of Transmission The organism is an inhabitant of the respiratory tract of pigs, and transmission occurs by direct nose-to-nose contact, which is the main form of transmission. Airborne transmission and fomites are less important. Mycoplasma can be transmitted over 1, 75, and 150 m, and recently aerosol has been seen to be transmitted over 9.2 km.12 M. hyopneumoniae was found to travel long distances from an infected experimentally infected group of pigs.13 Airborne transmission was suggested on 80% of farms where acute respiratory disease was present. No airborne organisms were found on farms without acute respiratory disease. There is no other known host for the organism, although infection and breakdown of closed pneumonia-free herds has occurred without any pig introductions. The number of organisms required for infection is very small, and the possibility of wind-borne infection has been suggested. Transmission is by the respiratory route and in infected herds occurs primarily from the sow to the suckling piglets. In a study of shedding of MH in different parities: gilts were 73% positive, parity 2 to 4 sows

were 42% positive, parity 6 to 7 sows 50% positive, and parity 8 to 11 sows were 6% positive. Generally, the nursery is considered the area where transmission occurs and infection spreads slowly. Within pen transmission measured by PCR is very slow. Animals can be PCR positive and not infectious for long periods of time and then can become very infectious up to 119 days, as has been recorded.11 There is therefore a nonlinear excretion of MH. It is thought that one infected nursery pig will infect on average one littermate. Boars can also infect sows when they are kept together in service areas, but in these areas the disease spreads slowly. The disease is also transmitted and exacerbated during the grouping and stress of pigs that occurs at weaning. Transmission can occur as early as 1 week of age, but usually it is not observed under 6 weeks of age.14 The highest clinical and pathologic incidence occurs in the postweaning and growing period, and in most herds this is maintained through the growing period to market age. The start of finishing is the critical point. Direct exposure (nose-to-nose contact) of pigs at 9 to 11 weeks of age to seropositive gilts results in seroconversion to the organism by 21 days and is most frequent by about 11weeks after exposure. The presence of gross lesions of pneumonia correlated with the seroconversion. Frequent coughing by infected, intensively reared pigs suggests that repeated aerosol exposure occurs and is an important natural mode of transmission of respiratory pathogens. There is general agreement that management and environmental conditions considerably influence the severity of the disease. The reinfection of enzootic pneumoniafree herds, recurrences or so-called breakdowns, occurs at a rate of about 3% of herds every 6 months. In a study of swine herds that had participated in the Pig Health Control Association Scheme in the United Kingdom, the close proximity of the uninfected herds to infected herds appeared to be the most important risk factor that could explain the introduction of the infection. The size of the herd, the density of the pig population in the area, the distance to the next road regularly used for transportation of pigs, and differences in topography were risk factors associated with reinfection. There was little evidence to indicate that unexplained breakdowns occurred in association with long-term latent infection in other herds from which animals had been imported. Clinical signs of enzootic pneumonia in these herds commonly did not occur for several months after the introduction of infected pigs. MH was not transmitted during a 20-week period when personnel weekly contacted susceptible pigs in a naïve herd after they had been in contact with pigs in an infected herd. A comprehensive herd specific

Diseases of the Swine Respiratory Tract

VetBooks.ir

prevention program is necessary to reduce transmission of disease caused by MH.15 Risk Factors The prevalence, incidence, and severity of pneumonia in swine herds are determined by interactions among infectious agents, the host, the environment, and management practices. This being said a large survey of the seroprevalence of MH in 50 finishing herds showed that there were no risk indicators. Each farm is an individual one, with the farm itself exerting a great effect. A recent study in suckling pigs at the age of weaning in the United Kingdom16 has suggested that an increase in the number of live pigs born alive was linked to a lower incidence of MH in suckling pigs at weaning. Grinding the piglets’ teeth also reduced the incidence of MH. A second dose of iron was also associated with a reduced level of MH. A low environmental temperature also produced an increase in the incidence of pneumonia. In a study of MH in coughing piglets (3-6 weeks of age) from 50 herds with endemic respiratory disease in Germany,17 it was found that MH was detected in the lavage fluid in 12.3% of the suckling piglets and 10.6% of the weaned piglets. The study showed that the detection of MH in young piglets is associated with one or two site production and inappropriate gilt acclimatization. In a study of nasal carriage in farmers using PCR it was found that 15% of farmers carried MH in their nose, but it is not possible to say that they were colonized.18 Animal Risk Factors Several factors such as breed, age, presence of diarrhea, the prevalence of atrophic rhinitis, birth weight, and weaning weight, have been examined as animal risk factors. In some herds, the risk of coughing and pneumonic lesions increased with increasing age of pigs within a herd. In a survey of two different groups of pigs slaughtered at different ages, the age-specific prevalence of pneumonic lesions was 2.7% in pigs less than 16 weeks of age at slaughter, but it increased rapidly when pigs were between 16 and 22 weeks of age at slaughter. Infection at an early age has a greater effect than infection later in life. Pigs coughing by 14 weeks of age were, on average, 6.2 to 6.9 kg lighter than those with onset of disease near market age. The highest seroconversion rate occurs between 3 to 4 months of age. In a recent experimental infection, 77.7% of the infected animals were still positive 185 days later, and 100% of the naturally infected animals were still infected at the same time. There may be selective differences in the colonization rates between litters. There may also be a sex effect on colonization. A longitudinal study of the diversity and dynamics of MH infection has been described.19 In a study of a large number of sows in northwestern Germany, it was found that the

risk of a sow being seropositive was increased in herds with two- or three-site production, when piglets were not vaccinated, when herds had 2-week farrowing intervals, and in herds without AI/AO management of the farrowing units. The lack of an acclimatization period for boars was also associated with the risk of a sow being seropositive.20 Immune Mechanisms Pigs that recover from experimentally induced enzootic pneumonia are resistant to subsequent challenge. The nature of the immunity induced by MH, whether serum or local antibody mediated, T-cell mediated, or a combination of these factors, is not clear. Based on lymphocyte transformation tests of experimentally infected pigs, it is possible that cell-mediated immunity correlates with protective immunity. The median half-life of passively acquired antibodies to MH is 16 days, the persistence of antibodies is related to the initial antibody concentration, and antibodies waned by 30 to 63 days after birth depending on initial concentration. It has been detected as late as 155 days of age. The titer of maternal antibodies is a major concern when pigs are vaccinated. The age of the piglet vaccinated is not the key factor. The level of the sow’s antibodies approximately 4 weeks prepartum are at their highest and similar to the levels in colostrum. Immunity is not conferred through colostral immunoglobulins, and thus piglets born from immune dams are susceptible to infection and clinical disease. No significant correlations have been found between the colostral antibody levels and the colonization status of the sows. The level of immunoglobulins to MH can be used to monitor infection in the herd. Pigs usually seroconvert to APP and then MH. Pigs raised under unfavorable conditions develop pneumonic lesions more frequently than pigs raised under better conditions, regardless of their immune status. Pigs vaccinated with inactivated MH organisms develop both a cell-mediated and humoral immune response, but they are not protected from challenge exposure by natural infection. Local immunity, particularly secretory IgA, is considered to be important in protection against mycoplasma infection. MH may suppress alveolar macrophage function, which may predispose the lung to secondary infection. The organism is very clever in evading the immune response, probably by changing the nature of the immune response to one that is less effective. To do this it causes the production of cytokines IL-1α and beta, IL-6, and TNF-α by macrophages and monocytes and induces local inflammation. This is essentially moving the immune response from a TH1 type response to a T-helper type 2 response. In an experimental study it was shown that PRRSV vaccine strain and the natural infection were able to induce T-regs in pigs

1065

naturally infected with MH. This suggests that the exacerbation of MH following PRRS may be attributable to the ability of PRRSV vaccination and viral infection to induce regulatory T cells.21 Pathogen Risk Factors MH adheres to the tracheal and bronchial mucosae and causes an extensive loss of cilia. An evaluation of the virulence factors of MH field isolates has been made. Environmental and Management   Risk Factors Pathogenesis MH colonizes the respiratory epithelium for a long time and produces a prolonged inflammatory response and suppresses and modulates the immune reactions. Little is as yet known about the virulence factors of MH. A wide variety of proteins are produced. Mycoplasma have the smallest genomes of organisms capable of separate existence. This genome encodes for several immunogenic proteins including a cytosolic protein p36 (which may have lactic dehydrogenase activity); membrane proteins P46, P65, and P74 (can produce neutralizing antibodies); and an adhesin P97. The P97 adhesin mediates adherence of MH to swine cilia. An adhesin-like protein (P110) composed of a P54 and 2 P28 units has also been found. Attachment is a complex process involving many gene products. A recent study of the total protein profile, glycoprotein profile, and size differences in the amplified PCR product of P97 adhesin genes suggests that there is an intraspecies variation in the MH population in the United States. Combination with the P102 adjacent gene allows the two proteins to contribute to cellular adherence.5,6 A highly immunogenic MH lipoprotein Mhp366 was identified by peptide–spot array,22 and this may be a useful method for detecting MH infections. The in vivo virulence of MH isolates does not correlate with in vitro adhesion assessed by a microtiter adherence assay.23 These observations suggest that mechanisms other than adherence may be responsible for observed differences in virulence. The Mycoplasma penetrate the mucus layer and attach to cilia. They appear only to attach to the cilia. They release calcium++ ions from the endoplasmic reticulum of the ciliated cells. As a result there is a clumping and a loss of cilia and excess production of mucus by goblet cells.24 This results in a dysfunction of mucociliary clearance. The secondary bacteria attach to the damaged epithelium. In experimental infections of tracheal explants with MH, it was shown that IL-10, IL-6, and IL-8 were produced.25,26 There is also a production of TNF-α and IL-127 and IL-18, but the production of IFN-γ is inhibited.28 These are possible mechanisms for the down-regulation of cell-mediated immunity

1066

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

that allows for the enhancement of the duration and severity of pneumonia with PRRSV and a mechanism to modulate the immune response. Macrophages have an impaired phagocytic activity after MH infection. MH also alters the function of B- and T-cell lymphocytes. The experimental inoculation of the J strain of MH into piglets causes gross pneumonic lesions that are detectable 7 to 10 days later. Moderately extensive pneumonia is present 6 weeks after inoculation, progressive recovery can be observed after 10 weeks, and residual lung lesions are detectable in a few pigs up to 37 weeks after inoculation. Experimental infections vary in their effects in clinical signs and pathology.29 MH causes peribronchiolar lymphoreticular hyperplasia and mononuclear accumulation in the lamina propria, which causes obliteration of the bronchial lumina. There is also perivascular lymphoid hyperplasia. The bronchial mucous glands undergo hypertrophy; there are increased numbers of polymorphonuclear cells in the bronchial lumina and macrophages in the alveoli. Lymphocytes, together with plasma cells and macrophages are responsible for the increase in the thickness of the interlobular septa as the disease progresses. The hyperplastic BALT (bronchial and bronchiolar associated lymphoid tissue) in enzootic pneumonia cases consisted of macrophages, dendritic cells, Tand B-lymphocytes, and IgG+ and IgA+ cells. In these aggregates CD4+ predominated over CD8+ cells. The cells in the BALT released IL-2, IL-4 TNF-α, and, to a lesser, extent IL-1α and β. IL-1α and TNF-α were also released in bronchoalveolar lavage fluids, and IL-6 and IL-8 were found in the mononuclear cells of the alveolar septa. Hyperplasia of type II alveolar epithelial cells is progressive as the disease becomes worse. Affected pigs cough persistently, show labored respiration and reduced exercise tolerance. The lesions are similar to those of chronic bronchitis. After infection, MH multiplies in tracheal and bronchial mucosae, adheres to the ciliated cells, and causes a cytopathic effect and exfoliation of epithelial cells. There is a significant increase in the gland/wall ratio and a decrease in the ratio of respiratory to expiratory resistance. The effects of this chronic pulmonary lesion have been the subject of considerable investigation. It is thought that the presence of mycoplasmal lesions uncomplicated by secondary bacterial infections has minimal effect on the production of the pig if the environmental conditions are suitable. The lesions will heal, and any loss in production from the initial infection will be regained by compensatory regrowth. Severe lesions or those accompanied by secondary bacterial bronchopneumonia and pleuritis will usually cause a significant decrease in average daily gain and feed efficiency. Secondary infection

with Pasteurella spp. results in acute episodes of toxemic bronchopneumonia and pleuritis. Dual infections are usually more severe than single infections. For example, SIV and MH together are more severe. A longitudinal study was made in four herds until slaughter. The percentage of pigs testing positive increased from 35% at 6 weeks to 96% at slaughter at 26 weeks. Within each herd only one distinct strain was detected19 and was present in the same animal for at least 12 weeks. The pulmonary and hematologic changes in experimental MH pneumonia cause no significant changes in heart rate, respiratory rate, and rectal temperature, even though at necropsy well-demarcated pulmonary lesions were present. There were several measurable changes in respiratory functions as a result of the atelectasis: partial occlusion of the bronchioles with exudate, localized pulmonary edema, and a reduction in oxygen perfusion to the alveoli leading to a decrease in the partial pressure of oxygen in the arterial blood. There are no remarkable changes in the hematology. The body weight gains are decreased compared with the control animals. The distribution of lesions is characteristic. They occur in the right middle lobe, the right cranial and left middle lobes, and the left cranial and diaphragmatic lobes, in that order of frequency. The differences in pathogenicity between high- and low-virulence isolates is associated with a faster in vitro growth, a raised capacity to multiply in the lungs, and the induction of a more severe inflammatory process.30 It has been shown recently that MH- derived lipid-associated proteins induce apoptosis in alveolar macrophages by increasing nitric oxide production, oxidative stress and caspase-3 activation.31 In a study to assess the duration of infection with MH 60 pigs were infected and studied until the population became negative on estimation of DNA in bronchial swabs. DNA was detected in 100% of the animals at 94 days postinfection, 615 at 214 days, and 0% at 254 days PI. Experimentally infected pigs transmitted to sentinels at 80 and 200 days post infection.32

CLINICAL FINDINGS

The appearance of clinical pneumonia depends on the number of organisms, their virulence, and the involvement of secondary agents. The more pathogenic strains induce more pneumonia.30 It is also influenced and made more severe by PCV233,34 and together with PRRSV is also more severe. A natural incubation period of 10 to 16 days is shortened to 5 to 12 days in experimental transmission. Two forms of the disease are described. In the relatively rare acute form, a severe outbreak may occur in a susceptible herd when the infection is first introduced. In such herds pigs of all ages are

susceptible and a morbidity of 100% may be experienced. Suckling piglets as young as 10 days of age have been infected. Acute respiratory distress with or without fever is characteristic and increased mortality may occur. The usual course of this form of the disease within a herd is usually about 3 months, after which it subsides to the more common chronic form. The chronic form of the disease is much more common and is the pattern seen in endemically infected herds. Young piglets are usually infected when they are 3 to 10 weeks of age, and clinical signs may be seen in suckling piglets. More commonly, the disease shows greatest clinical manifestation after weaning and in the growing period. The onset of clinical abnormality is insidious and coughing is the major manifestation. Initially only a few pigs within the group may show clinical abnormality, but then the incidence generally increases until coughing may be elicited from most pigs. It may disappear in 2 to 3 weeks or persist throughout the growing period. In affected herds, individual pigs may be heard to cough at any time, but coughing is most obvious at initial activity in the morning and at feeding time. Coughing may also be elicited by exercising the pigs around the pen, and it occurs with greater frequency in the period immediately following the exercise. A dry or crackling, hacking cough, which is usually repetitive, is characteristic. Respiratory embarrassment is rare and there is no fever or obvious inappetence. Subsequently there is retardation of growth that varies in severity between individuals so that uneven group size is common. Clinical disease becomes less obvious with increasing age and is rarely detected in the sow herd, although gilts and young sows frequently harbor MH.

CLINICAL PATHOLOGY

Raised haptoglobin levels have been found in pigs with lung conditions resembling Mycoplasma infection but not A. pleuropneumoniae–type lesions.35 Serologic Tests Serologic tests are best used to assess the herd status. All the three assays in use in the United States have excellent specificity, but the sensitivity is low, from 37% to 49%. The tests vary in their efficacy in different experimental infections.36 Serologic tests have included the CFT (low sensitivity), indirect hemagglutination test (good for early detection as it detects IgM), and the latex agglutination test. The unsatisfactory sensitivities and specificities of these tests led to the development of ELISA systems, DNA probe technology, and PCR to accurately diagnose enzootic pneumonia. The ELISAs detect all classes of IgG and are very sensitive, but they detect the onset of seroconversion rather than infection. An SIgA-ELISA has been developed for

Diseases of the Swine Respiratory Tract

VetBooks.ir

detecting secretory IgA from nasal swabs,37 and it is capable of detecting MH infection from MH vaccinated pigs. An ELISA using a commercially available antigen (Auspharm) is highly sensitive (95.6%) and specific (98.8%) for antibodies against MH when pig sera from commercial herds of known infection status were evaluated. An improved ELISA is also available, and the two ELISAs are able to distinguish populations of gross pathology-negative pigs in endemic herds from pigs in true specific pathogen-free (SPF) herds. Pigs from the former group have significantly higher ELISA activity with both tests and would represent recovered or exposed nondiseased pigs, or pigs with only histologic lesions in endemic herds. The ELISA is ideal for diagnostic laboratories and should obviate much of the need for culture and immunofluorescent histopathology, reducing the cost of diagnosis. The ELISA can also detect antibodies in the colostrum of sows with a high specificity. A recent study comparing three ELISAs has shown that the sensitivities of the tests were lower than previously reported especially for vaccinated animals. Animals within 21 days postinfection were also not easily detected. The blocking ELISA was the most sensitive. All three were highly specific. There is also a blocking ELISA against a p40 protein. Colostrum has also been used for the certification of freedom from MH but must be achieved during the first 2 hours after parturition. High-parity sows are a better source for the detection of antibodies. Detection of Organism For the highest level of accuracy in detecting the organism the use of a number of tests would be best. The organism can be detected in lung tissues by culture, immunofluorescence, PCR, and antigen-ELISA, and all have high sensitivity in the acute stages of pneumonia. A PCR-based assay can differentiate MH, M. flocculare, and M. hyorhinis and also detect low numbers of organisms. It can also be used on the bronchoalveolar lavage. The identification of the p36 and p46 protein genes has enabled them to be used in a PCR for MH, with a sensitivity of 86.6% and a specificity of 96.7%. Nested PCR is much better. There is a good correlation between the results of nested-PCR and histology. In situ hybridization shows MH on the surface of the epithelial cells, not in the cytoplasm, with an occasional signal in the cytoplasm of the alveolar and interstitial macrophages. A PCR38 had a diagnostic sensitivity of 97.3% and a specificity of 93.0%. Herd Certification The determination of the presence or absence of MH within a herd for certification purposes can be difficult and should be approached with caution. It should not be

based on a single examination procedure. It requires a surveillance system that combines regular farm visits and serologic, cultural, and tissue examination of selected pigs and of those sent to slaughter. The herd should be examined clinically for evidence of the disease, and the lungs from several shipments of pigs should be examined at the abattoir and subsequently histologically. There can be seasonal variation in the severity of lung lesions and at certain times market-age pigs may not have visible gross lesions, even though infection may be present in the herd. If doubt exists, the lungs of younger pigs, preferably clinically suspect pigs, or recently weaned pigs, should be examined after elective slaughter. The herd should also be examined for the presence of antibody to MH.

NECROPSY FINDINGS

Except in severe cases, the damage is confined to the cranial and middle lobes, which are clearly demarcated from the normal lung tissue. The lesions are commonly more severe in the right than in the left lung (simply because it is larger, has a larger supply of main-stem bronchi and a greater arterial supply). Plum-colored or grayish areas of lobular consolidation are evident. Enlarged, edematous bronchial lymph nodes are characteristic. In acute cases, there is intense edema and congestion of the lung and frothy exudate in the bronchi. When secondary invasion occurs, pleuritis and pericarditis are common, and there may be severe hepatization and congestion with a suppurative bronchopneumonia. Evaluation of the pneumonic lesions at slaughter has been used extensively for herd health monitoring. Scoring of the lesions is typically done on both lungs (the entire pluck). To overcome the logistical problems associated with examining entire plucks during the slaughtering procedure, an alternate system based on scoring the right lung only has been investigated. The overall right lung relative sensitivities for the detection of catarrhal pneumonia or chronic pleuritis were 81% and 72%, respectively. It is suggested that an evaluation of the right lung pathology is a useful alternative when the purpose of the survey is to demonstrate the presence or absence of lesions, or when scoring the severity of the lesion is the objective. The microscopic changes of enzootic pneumonia include lymphohistiocytic peribronchiolar cuffing with increased numbers of mononuclear leukocytes in the bronchial lamina propria. There is hyperplasia of the bronchiolar epithelium and filling of alveoli with macrophages, protein-rich fluid and small numbers of lymphocytes and plasma cells. Hyperplasia of type II alveolar epithelial cells occurs as the disease progresses. These histologic changes were most marked from 7 to 28 dpi coinciding with a

1067

significant increase in the immunohistochemical demonstration of Il-1α, IL-1β, IL-8, TNF-α and INF-γ, lymphoid markers CD4+ and CD8+, muramidase, and IgG and IgA.39 The lesions and immunohistochemical signals declined in intensity after 35 days. In one study, a definitive diagnosis of Mycoplasma pneumonia of swine was based on the demonstration of MH in lung sections using specific antisera or successful culture of the organism. Utilizing these techniques, it was found that up to 19% of grossly normal lungs may be infected with MH. Conversely, the organism could not be demonstrated in about 33% of the lungs of pigs from herds thought to be affected with MH pneumonia, even though typical gross lesions were present. The sensitivity of these techniques may be surpassed by newer PCR methods. The organism can also be detected in formalin-fixed paraffin-embedded porcine lung by the indirect immunoperoxidase test. The results of immunofluorescence tests performed on piglets with experimentally induced pneumonia revealed that MH organisms are located primarily on bronchial and bronchiolar epithelial surfaces of lungs with gross lesions of pneumonia. Fluorescence was most intense 4 to 6 weeks after infection and began to decrease at 8 to 12 weeks. This suggests a decrease in the number of MH in the more advanced stages of the disease. When assessing plucks at slaughter to determine the severity of pneumonia in a group, it must be remembered that in most instances the lesions observed represent a chronic, partially resolved disease process. Therefore the clinical effects of the infection may have caused a greater degree of respiratory compromise than is apparent at slaughter. In a recent study the histopathology of lungs in slaughter pigs vaccinated with different vaccines has been described.40 Lung lesion scores and MH loads differed widely between the three different vaccine groups but were correlated with each other.

DIAGNOSIS

Typical epidemiology and a dry hacking cough are suggestive of MH. Typical lesions need to be investigated at the margins of the lesions and culture attempted. Recently farms have been described that have more than 1 strain of MH.1 Tracheal bronchial swabbing associated with RT-PCR could be an accurate diagnostic method.41 The most sensitive sampling methods for detecting MH in live, naturally infected pigs were tracheobronchial swabbing or washing compared with oropharyngeal brushing and nasal swabbing.42 Samples for Confirmation of Diagnosis • Touch preparations using Giemsa stained slides have been used.

1068

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

• Histology—formalin-fixed lung (LM, IHC). Simple histopathology may not always indicate MH infection. For example, Aujeszky’s disease together with P. multocida may be difficult to differentiate from MH. Lesions may be characteristic but not pathognomonic. Detection in lung tissue is either by FA or IHC, and these are rapid and cheap and more often used than ISH. The more fresh the material or immediately fixed material gives better results. In experimentally infected pigs MH could be reisolated from liver and spleen of experimentally infected pigs and contact pigs.43 Indirect immunofluorescence (IF) and indirect immunoperoxidase (IHC) for MH in tissues are extremely useful. However, IF has a lack of sensitivity and IHC is time consuming and expensive. Mycoplasmology lung (MCULT, FAT, PCR). Isolation of MH is complicated by the overgrowth that occurs from M. hyorhinis and M. flocculare. The organism is fastidious, and 4 to 8 weeks are sometimes needed for growth. It also requires specialized media, including swine serum. For these two reasons it is not so commonly used now. Many animals that are culture positive do not have gross or microscopic lesions. PCRs have become a sensitive and specific method for identifying MH.38,41 Lung tissue, bronchial swabs, or bronchial washings are the best sites. Nested PCRs raise the sensitivity and may detect as few as four to five organisms. A real-time TaqMan PCR that simultaneously detected the proteins P46, P97, and P102 has been designed44 that can detect 108 Mycoplasma per pig. In addition, a multiplex PCR has been developed that can be used on culture broth for several mycoplasma.45 A number of RT-PCRs have been developed that allow quantification.44,46 The PCR can be used as a one-step test but is not good for nasal swabs. The nested PCR can be used for these, but it does tend to produce some false positives. Correct samples give a better diagnosis. Samples from lavage and tracheobronchial sites were the best for nested PCR, and lung tissue and nasal swabs are not the most reliable.43

TREATMENT

There is no effective treatment to eliminate infection with MH, although the severity of the clinical disease may be reduced. Isolates of the organism from the United States were susceptible to lincomycin-spectinomycin, tylosin, and oxytetracycline. Isolates from the United Kingdom were susceptible to doxycycline and oxy­ tetracycline. Doxycycline, a semisynthetic tetracycline, has a greater antimicrobial activity, is better absorbed orally and is more widely distributed in tissues than the

first-generation tetracyclines (oxytetracycline, tetracycline, and chlortetracycline). Tetracyclines given as a preventative in-feed are more effective than giving tetracyclines once clinical signs of coughing have started.47 This is particularly true when using the drug around times of stress and acquisition of the organisms (ie, in the nursery and at weaning). A recent study has shown that CTC when administered at the onset of clinical signs via the feed at a dosage of 500 ppm during two alternate weeks was able to decrease the prevalence of pneumonia lesions and numerically reduced the performance losses and clinical signs.48 In some early studies, a mixture of tylosin tartrate at a dose of 50 mg/kg BW and tiamulin at 10 mg/kg BW orally daily for 10 days significantly reduced the pulmonary lesions associated with the experimental disease. However, the use of 60, 120, or 180 mg of tiamulin per liter of drinking water for 10 days was not effective in suppressing the lesions of experimentally induced MH pneumonia or infection in disease-free pigs. The newer fluoroquinolones have good in vitro activity against MH and exhibit superior activity to tylosin, tiamulin, oxytetracycline, and gentamicin. Ciprofloxacin is particularly active against MH. Tilmicosin is particularly effective because it appears to prevent the attachment of MH to the surface of the epithelial cells. Tetracyclines will either prevent transmission or suppress lesion formation in experimental pigs but the levels required are high and in an infected herd continuous administration would be necessary, which would be uneconomic. Treatment is generally restricted to individual pigs showing acute respiratory distress as a result of a severe infection or secondary invaders. Broad-spectrum antimicrobials are used, usually tetracyclines, but the response is only moderately good. The occurrence of severe signs within a group of pigs may necessitate treatment. Tetracyclines, tylosin, or spiramycin fed at 200 mg/kg feed for 5 to 10 days is recommended. A combination of 300 g of oxytetracycline and 30 g of tiamulin per ton of finished feed fed for 2 to 3 days/week over a 16-month period has been used to reduce the incidence of enzootic pneumonia in a large herd. Lung lesions were reduced, average daily gain increased, and efficiency of feed conversion increased, with an overall increase in profitability. Valnemulin may prove to be effective in the treatment of enzootic pneumonia. There is a higher susceptibility to valnemulin and tiamulin when used in conjunction with doxycycline as a treatment. Tulathromycin administered as a single injection at a standard dosage of 2.5 mg/kg is effective in the treatment of swine pneumonia associated with mycoplasmosis.

Oral florfenicol feed supplementation (20 g/ton) reduces the effects of MH infection.49 There is no evidence for resistance to lincomycin/spectinomycin, oxytetracycline, doxycycline, gentamicin, flufenicol, and tiamulin. There is evidence for some resistance from the field to tetracyclines, macroloides, linosamides, and fluoroquinolones.50,51

CONTROL

Control strategies have been reviewed.52 In all cases recommended management procedures such as all in/all out pig flow, medicated and segregated early weaning, and multisite operations further facilitate control of respiratory disease. MH infects only pigs and transmission requires close pig-to-pig contact. If transmission can be prevented, it is possible to limit or even eradicate the disease from a herd. There are thus two levels at which control can be practiced: (1) Complete eradication of the disease or (2) Controlling the disease and its effects at a low level. The principles of control of MH include the following strategies: • Regular inspection of the herd for clinical evidence of disease and slaughter checks of lungs • Rigorous biosecurity of animals being introduced into the herd and control of visitors • Provision of adequate environmental conditions, including air quality, ventilation, temperature control, and stocking density • The use of the all-in, all-out system of production in which groups of pigs by age or stage of production are moved through the herd from the gestation barn, farrowing barn, nursery rooms, and finishing units as groups and the pens previously occupied are cleaned, disinfected, and left vacant for several days before animals are reintroduced. Because most infection is believed to occur between 4 to 12 weeks, nursery depopulation has become an effective way of controlling the infection in nursery pigs. Control by Eradication Control by eradication is the most satisfactory and is probably mandatory for large breeding companies, herds supplying replacement stock to other herds, and for large intensive farrow-to-finish enterprises. It is based on the principle that the source of infection for the young pig is the gilt or the sow and this chain of infection must be interrupted to prevent infection. In the past, the 10-month cutoff point has been used in eradication programs, but in view of the colonization studies, this may be too soon. This is especially so in off-site production systems where the time of infection is delayed.

Diseases of the Swine Respiratory Tract

VetBooks.ir

There are three different principle methods. First, there is total depopulation followed by restocking with noninfected stock (Danish SPF system). Second, the test and removal of all positives and inconclusives. Third, complete eradication without total depopulation and restocking. Eradication without restocking has been described, and here the secret was to wait until farrowing finished, then vaccinate all sows and treat with tiamulin at 6 mg/kg daily for 3 weeks and then monitor with blood tests.

litter is kept as a separate unit. The litter is inspected clinically at regular intervals, and subsequently a proportion of the litter, usually excess males and gilts undesirable for breeding, will be examined at slaughter for evidence of pneumonia. Any litters with clinical, pathologic, or laboratory evidence of pneumonia are eliminated from the program. Litters that pass inspection are kept for repopulation of the herd. Because of the difficulties in detecting carrier pigs without lesions, eradication by methods using these principles frequently fails.

Specific Pathogen-Free or Minimal-Disease Pigs Several methods of eradication have been attempted, but the most satisfactory is repopulation with specific-pathogen-free (SPF) pigs. The principle underlying this method is that the piglet in utero is free of infection with MH. If it is taken from the uterus at term by suitable sterile hysterectomy or hysterotomy techniques and reared artificially in an environment free of pigs, it will remain free of this infection. In practice this has been carried out in special units, and the piglets have been subsequently used to repopulate existing farms where all pigs have been removed 30 days before the introduction of the SPF pigs and a thorough cleaning program completed. This method was initially developed for the control of MH and atrophic rhinitis. Moreover, if suitable precautions are taken and if the piglets are used to populate new units that have had no previous exposure to pigs, then freedom from other important diseases such as internal and external parasitism, leptospirosis, brucellosis, swine dysentery, and others can be achieved. The progeny of these primary SPF herds can subsequently be used to repopulate other or secondary SPF herds known as minimal disease pigs. Because of the cost and technical difficulty of this method, other methods of eradicating MH have been attempted, but they are generally less satisfactory and have a higher failure rate. These include “snatching” of pigs at birth and isolated farrowing. In the former the piglets are caught and removed from the sow immediately at birth and reared as previously described or foster-suckled on SPF sows in another environment. Although MH may be eliminated by this method, fecal contamination during parturition of the vulva and vagina and consequently of the piglet is common, and this method is less satisfactory for disease control than removal by hysterectomy.

Minimal Disease Herds Minimal disease herds have been established in most countries with significant pig populations, either by breeding companies or private purebred breeders. As a result, in most countries there is a nucleus of MH-free stock. The establishment of primary SPF herds is technically difficult and very costly and should not be undertaken lightly. There is also a considerable delay in cash flow between the time of initial population and buildup of herd numbers to the time when significant numbers of pigs are available for sale. If eradication by repopulation is intended, it is preferable to purchase pregnant gilts from established primary SPF herds unless the maintenance of existing genetic lines dictates otherwise. Before recommending eradication by this method it is essential that the pig owner understands the principles of this method of control and the restrictions that will need to apply if it is to be successful. Farrow-to-finish enterprises established by this method should be run as closed herds, and if further genetic material is required it should be introduced by hysterectomy techniques or by purchase from the initial source herd. The use of artificial insemination is an alternate method; however, isolation of MH from semen has been recorded. The problem of certifying and maintaining herds free of MH is a major task.

Isolated Farrowing Isolated farrowing techniques have proved successful in small herds but have a high failure rate when practiced on a large scale. Older sows believed to be free of infection are farrowed in isolation in individual pens erected outside on pasture and each sow and

Reinfection of Herds Reinfection of MH-free pig herds occurs despite high standards of isolation and strict precautions when complete protective clothing and showering routines are required for all visitors entering the unit. All visitors are debarred entry if they have been to a possible source of infection during the previous 48 hours and even up to 7 days. Also, the majority of breakdowns occur in herds that have not imported infected stock recently. In reinfected herds that imported stock there was no concurrent evidence of breakdown in the parent herds, which supported the contention that the importation of infected pigs was an unlikely source of the infection. An epidemiologic investigation of these reinfections suggests that close proximity of uninfected herds to infected herds may be an important factor. The organism does not

1069

survive for more than a few days under dry conditions; however, it can survive in diluted tap water and rainwater for 2 to 3 weeks, and it has been suggested that the organism may be transported in moist air and that airborne infection between piggeries is a possible method of transmission. In Switzerland, 107 farms were reinfected of the 3983 that were eradicated during the period 1996 to 1999 (2.6%). The significance of known risk factors such as farm size, high density of pigs, and farm type was confirmed in this analysis. Some preliminary estimates of risk indices based on the proximity of other pig units has indicated that the most important factor was the reciprocal of the square of distance to the nearest other unit. The crucial distance for maximum survival was about 3.2 km. A breakdown was described recently in which a whole variety of measures were included in an attempt to control the disease. Antimicrobial Prophylaxis Eradication has also been attempted by antimicrobial treatment of newborn piglets with oxytetracycline on days 1, 7, and 14, which were weaned on day 14 and moved to offsite nursery. This is known as a low-cost modified medicated-early-weaning program. This can be followed by serologic testing of the breeding herd and culling of positive reactors. Control by vaccination on the one hand and by the use of tilmicosin on the other produced similar results when measured by serologic results and the prevalence of macroscopic lung lesions. Lincocin with or without vaccination considerably improves the growth and performance. Doxycycline in the feed at 11 mg/kg BW is effective in controlling pneumonia caused by P. multocida and MH in feeder pigs. Low-Level Disease The alternative to eradication is to limit the effects of the disease in those herds where eradication is either not desirable or feasible. The effects of the disease are generally less severe in nonintensive rearing situations, in small herds where individual litters are reared separately, and where litters from older sows can be reared separately from other pigs. Where litters are grouped at weaning, a low stocking density with less than 25 pigs in initial pen groups and 100 pigs in a common airspace may also reduce the severity of the disease. Temperature, humidity and ventilation also have an important influence on the disease. It is possible to determine an optimal air temperature zone for growing-finishing pigs based on the measurement of behavioral and health-related problems. They are interrelated with stocking density and housing. The subject is too broad for treatment here, and the requirements for pigs at different ages and under different housing situations may be found in standard texts on pig

1070

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

housing and production. The environmental risk factors associated with the incidence of MH should be assessed in each circumstance. Some important environmental variables that should be assessed and modified include the following: • Number of pigs per shed • Number of pigs per pen • Airspace per pig • Floor space per pig • Cleaning and disinfection techniques used • Number of air changes per hour • Waste disposal system • Number of temperature fluctuations in a 24-hour period • Direction of the flow of air in the building • Concentrations of ammonia and hydrogen sulfide in the building • Dust levels • Feeding and watering systems • Whether or not the all-in, all-out system is being used effectively Medication of Breeding Stock The original medicated early weaning program was based on medication of the sows with tiamulin at the time of farrowing and the early weaning of the piglets to an off-site location. A variation of this method is to prevent the spread of infection by the following means: • Isolation of the breeding stock • Strategic antimicrobial medication of the breeding stock • Reintroduction of the breeding stock to the original but empty and disinfected gestation barn • Separate rearing of the piglets before and after initiating the program • Regulation of flow of animals through the herd. Farrowing barns are emptied out when possible and cleaned, disinfected, and left empty. After weaning their piglets, sows are transferred to the dry sow barns. Sows about to farrow are treated with tiamulin and moved to the farrowing barn. Medication and vaccination was used to eradicate MH without total depopulation.53 Source of Feeder Pigs Where possible the purchase of weaners or pigs for finishing units should be from herds free of the disease or from a single source. Purchase through saleyards or the purchase of coughing or uneven litters is not advisable. When pigs from infected herds are purchased it may be necessary to medicate the feed prophylactically with one of the tetracycline group of antibiotics or tylosin or spiramycin at 100 to 200 mg/kg of feed for a 2-week period after introduction. Medication of the feed of finishing pigs with tiamulin at 20 and 30 mg/kg of feed over an 8-week period on farms with histories of severe

complicated enzootic pneumonia resulted in improved weight gains and feed efficiency, but the extent and severity of the lung lesions did not change. The level of 30 mg/kg in the feed was superior to the level of 20 mg/kg. Tiamulin at 100 mg/kg combined with chlortetracycline at 300 mg/kg of feed for 7 days was effective in herds with a history of MH complicated by the presence of P. multocida and Actinobacillus pleuropneumoniae. Introduced pigs should be isolated from the rest of the herd and preferably they should be reared as a batch through a house on the all-in, all-out system. A high stocking density should be avoided and internal parasites should be controlled. Vaccination A general observation was that Ascaris sum infection affected the response to vaccination for MH.54 Vaccination reduces the macrophage infiltration in bronchus associated lymphoid tissue infected with a virulent MH strain.55 In the same study MH was reduced in the lungs in the vaccinated pigs, and the high-virulence strain was inhibited more than the low virulence strain. Vaccination significantly reduces clinical signs macroscopic and microscopic lung lesions especially infected with avirulent strain.56 The effect was less pronounced with a less virulent strain. Vaccination does not, however, reduce the transmission to other pigs.57 MH vaccines are generally bacterins consisting of outer membrane proteins or whole organisms. The vaccines give little protection against initial infection and often incomplete protection against clinical pneumonia. The vaccines produce a TH1 response and also IgA and IgG in the lavage fluids. Natural maternal antibodies do not seem to inhibit vaccination, but vaccination of sows may inhibit subsequent immunity. Vaccination with killed MH induces protection in pigs against experimental challenge exposure with the organism. A cost-benefit analysis shows that the vaccination is economically beneficial. The relationships between maternally derived antibodies, age, and other factors in vaccine response have been discussed. Intranasal vaccination of attenuated MH adjuvanted with bacterial DNA may be effective in evoking the local cellular and humoral response and the systemic immune response.58 A killed MH vaccine evaluated in a single herd reduced the prevalence of pneumonic lesions in slaughter pigs from 69% to 36% and the prevalence of pleuritis from 20% to 13%. There was a small decrease in the number of days to market. It usually results in a 2% to 8% increase in daily gain. The mortality rate is usually only better numerically. Feed conversion efficiency increases by about 2% to 3%. Other limited studies indicate that vaccination can reduce the severity and prevalence of lung lesions detected at

slaughter (4%–6% compared with 12% in controls). It improves feed efficiency and increases average daily gain during the finishing period. In other studies the average daily gain was not improved. Under experimental conditions the transmission in nursery pigs was only numerically lower in vaccinated pigs and the vaccination does not prevent the establishment of MH in the lung.59 Vaccination of piglets improved pulmonary health, but vaccination of sows alone did not prove to be sufficient.60 Vaccination of sows against MH reduced the prevalence of positive piglets at weaning and could be used to control MH infections as judged by a nested PCR. PRRS vaccination does not interfere with MH vaccination. Needle-less intradermal vaccination has also been described. Double-vaccinated pigs show a lower percentage of MH-compatible gross lesions and a lower MH prevalence in the URT compared with single vaccinated animals.61 Both dual and single injection vaccines are available, but the protection obtained is similar. The single dose vaccine gives protection for up to 23 weeks. The level of protection will probably last 4 months. Vaccination is economically attractive. DNA vaccination using a p42 heat-stable protein gene has also been used, and this induces rises in IL-2, IL-4, and IFN-γ, which indicates that it induces both a Th1 and a Th2 response. Vaccination for mycoplasma generally induces local mucosal immunity, humoral and cellular immunity. A recent study has shown that inactivated vaccine produced both systemic and mucosal cellular and humoral immune responses.62 It appears to prime the immune response, but this may not become fully operational until natural exposure takes place.63 Sow vaccination strategies are still undergoing study but it has been shown that the severity of the pneumonia in piglets born to vaccinated sows was reduced.64 It increased the percentage of seropositive sows and piglets at weaning but did not affect the sow or piglet colonization. Maternal antibodies do not interfere with vaccination unless they are very high. PRRSV infection may reduce the response to vaccination, but this may depend on the strains of both agents. In a study there were no significant differences between the protective efficacy of a combined PRRSV/MH vaccine and the two single vaccines.65 Intradermal vaccination was successful in reducing lesions by 10.4% compared with controls, and 6% in the intramuscular injection group. Intradermal vaccination afforded greater protection especially with regard to morbidity, lung lesion, and pleuritis scores.66 Subunit vaccines may be developed in the future, and other immunodominant antigens other than P97 should be taken into account.67

Diseases of the Swine Respiratory Tract

FURTHER READING

VetBooks.ir

Desrosiers R. A review of some aspects of the epidemiology, diagnosis and control of M. hyopneumoniae infections. J Sw Hlth Prod. 2001;9:233-237.

62. Marchioro SB, et al. Vaccine. 2013;31:1305. 63. Martelli P, et al. J Vet Med B. 2006;53:229. 64. Sibila M, et al. Vet Microbiol. 2008;127:165. 65. Drexler CS, et al. Vet Rec. 2010;166:70. 66. Tassis PD, et al. Vet Rec. 2012;170:261. 67. Okamba FR, et al. Vaccine. 2010;28:4802.

REFERENCES

1. Mayor D, et al. Vet Res. 2007;38:391. 2. Mayor D, et al. Vet Microbiol. 2008;127:63. 3. Stakenborg T, et al. Vet Res Commun. 2006;30:239. 4. Villareal I, et al. Vaccine. 2009;27:1875. 5. Burnett TA, et al. Mol Microbiol. 2006;60:669. 6. Wilton J, et al. Mol Microbiol. 2009;71:566. 7. Pinto PM, et al. Vet Microbiol. 2007;121:83. 8. Kuhnert p, et al. Vet Microbiol. 2011;152:191. 9. Sanchez-Vazquez MJ, et al. Pig J. 2010;63:25. 10. Fano E, et al. Can J Vet Res. 2007;71:195. 11. Pieters M, et al. Can J Vet Res. 2010;74:157. 12. Sanchez-Vazquez MJ, et al. Pig J. 2010;63:25. 13. Dee S, et al. Vet Res. 2009;40:30. 14. Sibila M, et al. Vet Microbiol. 2007;121:352. 15. Nathues H, et al. Acta Vet Scand. 2013;55:30. 16. Nathues H, et al. Acta Vet Scand. 2013;55:44. 17. Moorkamp L, et al. Transbound Emerg Dis. 2009;56:54. 18. Nathues H, et al. Vet Rec. 2012;170:623. 19. Vranckx K, et al. Vet Microbiol. 2012;156:315. 20. Beilage Eg, et al. Prev Vet Med. 2009;88:259. 21. LeRoith T, et al. Vet Immunol Immunopathol. 2011;140:312. 22. Meens J, et al. Vet Microbiol. 2010;142:293. 23. Calus D, et al. J Appl Microbiol. 2009;106:1951. 24. Kim CH, et al. Vet J. 2012;192:120. 25. Choi C, et al. J Comp Pathol. 2006;134:40. 26. Lorenzo H, et al. Vet Immunol Immunopathol. 2006;109:199. 27. Ahn KK, et al. J Vet Med Sci. 2009;71:441. 28. Muneta Y, et al. J Interferon Cytokine Res. 2006;26:637. 29. Woolley LK, et al. Vet Microbiol. 2012;161:186. 30. Meyns T, et al. Vet Microbiol. 2007;120:87. 31. Bai F, et al. Vet Immunol Immunopathol. 2013;155:155. 32. Pieters M, et al. Vet Microbiol. 2009;134:261. 33. Dorr PM, et al. J Am Vet Med Assoc. 2007;230:244. 34. Wellenberg GJ, et al. Vet Microbiol. 2010;142:217. 35. Amory JR, et al. Res Vet Sci. 2007;83:428. 36. Strait EL, et al. J Clin Microbiol. 2008;46:2491. 37. Feng Z-X, et al. Vet Microbiol. 2010;143:410. 38. Cai HY, et al. J Vet Diag Invest. 2007;19:91. 39. Redondo E, et al. J Comp Path. 2009;140:260. 40. Hillen S, et al. Prev Vet Med. 2014;doi:.org/10.1016/ jprevetmed.2013.12.012. 41. Fablet C, et al. Vet Microbiol. 2010;143:238. 42. Fablet C, et al. Epidem Sante Anim. 2012;61:149. 43. Marois C, et al. Vet Microbiol. 2007;120:96. 44. Marois C, et al. J Appl Microbiol. 2010;108:1523. 45. Stakenborg T, et al. J Microb Methods. 2006;66:263. 46. Strait EL, et al. J Swine Hlth Prod. 2008;16:200. 47. Thacker B, et al. J Swine Hlth Prod. 2006;14:140. 48. Del Pozo Sacristan R, et al. Vet Rec. 2012;171:645. 49. Ciprian A, et al. Res Vet Sci. 2012;92:191. 50. Le Carrou J, et al. Antimicrob Agents Chemother. 2006;50:1959. 51. Vicca J, et al. Microb Drug Resist. 2007;13:166. 52. Maes D, et al. Vet Microbiol. 2008;149:41. 53. Heinonen M, et al. Vet J. 2011;188:110. 54. Steenhard NR, et al. Vaccine. 2009;27:5161. 55. Vranckx K, et al. BMC Vet Res. 2012;8:24. 56. Villareal I, et al. Vaccine. 2011;29:1731. 57. Villareal I, et al. Vet J. 2011;188:48. 58. Li Y, et al. Vaccine. 2012;30:2153. 59. Meyns T, et al. Vaccine. 2006;24:7081. 60. Strauss C, et al. Tierartzl Prax. 2007;35:283. 61. Sibila M, et al. Vet Microbiol. 2007;122:97.

PORCINE RESPIRATORY DISEASE COMPLEX AND MYCOPLASMAL PNEUMONIA OF PIGS Mycoplasma hyopneumoniae (MH) is a significant contributor to the porcine respiratory disease complex (PRDC), together with PRRS, PCV2, SIV, and secondary bacterial agents such as Pasteurella multocida1(PM), Actinobacillus pleuropneumoniae (APP) and H parasuis (HPS), E. coli, Klebsiella, Trueperella pyogenes, Bordetella bronchiseptica, streptococci, and staphylococci.2-6 In the study in Denmark,3 five bacterial species, five viruses, and two Mycoplasma species were found in different combinations. The study in Germany4 found that among a variety of pathogens, PCV2 and alpha-hemolytic streptococci were most frequently detected. There were also more associations between the organisms in clinical cases than in the healthy pigs. Porcine respiratory disease complex is a better name for what was once called enzootic pneumonia. This term really means pneumonia that occurs naturally in the population and includes a complex of many bacterial and viral agents with the occasional addition of parasites and protozoa. Some primary pathogens such as MH and APP are not usually isolated from healthy pigs and may be responsible for subclinical infections. A Danish study found that Actinomyces hyovaginalis was a common isolate from pyemic lungs in pigs. The authors did a study in the 1960s involving full viral, bacteriologic, and environmental and management analyses, which showed that each farm was an individual with its own set of variables, and that the only significant factor was that MH was associated with clinical disease and economic loss. Simultaneous occurrence of Aujeszky’s disease does increase the severity of acute mycoplasmal pneumonia. The jury is still out as to whether TTV has a role to play in PRDC.7 In a recent study, lipoteichoic acid from Staphylococcus aureus exacerbated respiratory disease in porcine-coronavirus infected pigs.8 Normally, the bacteria live in symbiosis with the host. The three major enzootic pig viruses (PRRS, PCV2, and SIV)9-13 destabilize the situation through direct pathologic effects or disturbances of the immune system. This complex is characterized by slow growth, decreased food conversion efficiency, anorexia, fever, cough, and dyspnea in grower finisher pigs typically around 16 to 22 weeks of age. It corresponds to what was originally called enzootic pneumonia.

1071

ETIOLOGY Some of the bacteria live happily in the upper respiratory tract, for example, Bordetella bronchiseptica (BB), some strains of Hemophilus parasuis (HPS), and M. flocculare, and, M. hyorhinis.14 Other organisms are inhaled directly or more likely introduced by noseto-nose contact (MH) or even aerosols, whereas others flare up in times of stress from small numbers normally harbored in the nasopharynx and tonsils, such as APP and PM. There is variation in the strains of many of these organisms and this determines the outcome of infection in many cases. Similarly, there may be breed dispositions to some of the agents.15,16 The presence of PRRS, P. multocida, H. parasius, M. hyorhinis, or S. suis correlated with a higher probability of also finding MH.17

EPIDEMIOLOGY

The combination of pathogens involved in the respiratory disease complex is legendary and varies from country to country, region to region, and even farm to farm.18 When a new agent enters the field (e.g., the pandemic SIV200919-23 or Torque teno virus), then the position becomes even more complicated until the population at large becomes immune. Secondary bacterial pneumonia can be a significant cause of mortality in the weaningto-market period. Some of the risk factors for pleuritis and cranioventral pneumonia have recently been reviewed.24,25 The relationships between the infectious and noninfectious factors in PRDC have been reviewed.26 Atrophic rhinitis may also be present along with enzootic pneumonia, and the two diseases in combination may have a greater economic effect than either disease alone. When outbreaks of respiratory disease in pigs occur, they are frequently the result of complex interactions between many agents. The importance of MH is not only its effect as a primary pathogen but also its ability to act synergistically with other infecting agents to produce significant respiratory disease. MH causes a mild pneumonia, whereas P. multocida is not pathogenic alone but aggravates the pneumonia initiated by the former pathogen. The epidemiologic associations between MH and Actinobacillus pleuropneumoniae antibody titers, and lung lesions in pigs at slaughter have been examined. Only titers to the Mycoplasma pneumonia were associated with lesions. The extent of the lesions produced by MH in PRDC may be influenced by other contributing factors to account for the variations in severity of lesions. Concurrent infection with lungworm, migrating ascarids, and an adenovirus has resulted in lesions of greater severity and secondary invasion of pneumonic lesions by Pasteurellae, Streptococci, Mycoplasma, and Bordetella bronchiseptica;

1072

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Klebsiella pneumoniae is very common and largely influences the outcome of the disease in individual pigs. In some abattoir surveys of lungs, P. multocida can be cultured from 16% of normal lungs and from 55% of lungs with lesions resembling those of enzootic pneumonia. P. multocida and Haemophilus spp. may also be found in conjunction with MH in the lungs of slaughter-weight swine affected with pneumonia and examined at the abattoir. Those lungs with both MH and P. multocida had the most macroscopic pneumonia, and those lungs with either of the agents alone had much less pneumonia. MH renders the lungs susceptible to P. multocida colonization and infection. Along with MH, other Mycoplasma species, such as M. hyorhinis, Acholeplasma granularum and Acholeplasma laidlawii, have been isolated from the lungs of pigs at slaughter, but their significance is unclear. MH and Mycoplasma hyorhinis have been isolated from 30% and 50% of pneumonic lungs, respectively, from pigs examined at slaughter. MH was also isolated from 12% of lungs with no gross lesions of pneumonia. In a survey in Norway, MH, P. multocida and M. hyorhinis were detected in 83%, 43%, and 37% of the pneumonic lungs respectively. Most of the macroscopic pneumonia—up to 25%—occurred in lungs with all three pathogens. M. flocculare was the most frequently isolated organism in the nonpneumonic lungs. MH potentiates the severity of PCV2associated lung and lymphoid lesions and increases the amount and perhaps the presence of PCV2 antigen. It also increases the incidence of PMWS in pigs. Several environmental and management factors are associated with a high prevalence of pneumonic lesions at slaughter. They include continuous versus all-in, all-out production, open herds, large temperature fluctuations, semisolid pen partitions, and large numbers of pigs in a common airspace. These factors may operate individually or in combination synergistically. Housing pigs in a clean, isolated, disease-free and low-stress environment positively influences the health of pigs. Complex animal production systems in the industrialized world have been reviewed.27 The primary and secondary pathogens of the disease produce their most detrimental economic effects and the highest level of morbidity and mortality during the finishing period when the economics of production necessitate indoor housing and intensification. Four main groups of environmental factors that contribute to high levels of clinical disease and lesions at slaughter include: 1. Meteorologic 2. Population and social 3. Management 4. Airborne pollution

Meteorological factors include wide fluctuations in the temperature indoors, wide variations in relative humidity, irregular ventilation rates, and winter housing. However, experimentally, elevated concentrations of ammonia and fluctuating ambient temperature did not influence the severity of the pneumonia or its effect growth rate. The noninfectious factors associated with pneumonia and pleuritis in slaughtered pigs in 143 farrow-to-finish farms in France were analyzed.28,29 Population factors that contribute to an increased prevalence of pneumonia are increasing herd size, increased population density, and decreased airspace and floor space per pig. All management practices influence the microclimate, and the quality of housing and management influences the incidence of pneumonic lesions at slaughter. Larger-than-average swine farms milling their own feed and with characteristics of modern buildings (mechanized inlets, slatted floors) and in close proximity to other farms tend to have a higher risk of enzootic pneumonia. Extensively housed pigs with aboveaverage pen space and air volume have a reduced prevalence of enzootic pneumonia lesions. Management factors associated with enzootic pneumonia include family farms that feed pigs on the floor and feeder barns that obtain pigs from multiple sources compared with those with good facilities and where the pigs originate directly from breeding units. The disease is a particular problem in continuous-flow herds. In pigs reared in all-in, all-out groups in the farrowing house, nursery, and growing-finishing unit, any Mycoplasma transmitted from sows to pigs or between pigs do not necessarily result in clinical signs or lesions of pneumonia. Pigs reared in all-in, all-out systems do not have lesions or minimal lesions at slaughter and gained at a faster rate than litter-mate pigs reared in a continuous system. Risk factors in suboptimal housing in Australia were described.30 In small herds, the factors commonly associated with a high prevalence of enzootic pneumonia were larger numbers of pigs per pen section, larger group sizes, and drafty farrowing and weaner accommodation. A study of housing density on species diversity and number of airborne microorganisms at fattening facilities has shown that the total number of bacteria and fungi did not exceed 104 and 103 CFU per m3 respectively. The number of organisms correlated with housing density. The most numerous were gram-positive bacteria and then gramnegative bacteria and fungi.31 Airborne pollution in pig houses is thought to contribute to an increased incidence of clinical disease and prevalence of lesions at slaughter.32 The pollutants include microorganisms, endotoxic cell wall constituents, and ammonia.29 Ammonia is the

most important because it is a powerful ciliotoxic agent in its own right before determining its effects on microorganisms. Toxic levels of ammonia, high concentrations of aerial dust, and high colony counts of aerial bacteria may contribute to an increased incidence and prevalence of pneumonia, but these factors have not been quantified and are commonly based on subjective evaluations by the observer. A large study of 960 pigs has shown that there are no influences of ammonia or dust on the respiratory health of pigs. Environmental air contaminants such as dust, ammonia, carbon dioxide, and microbes in swine barns measured over a period of 12 months were associated with lesions of pneumonia and pleuritis at slaughter. In a study of experimentally infected animals, it was found that 6/114 long-distance samples were positive for MH. Three samples collected at 3.5, 6.8, and 9.2 km from the herd of origin were infectious.33 In large herds, factors associated with a high prevalence were higher pen stocking rate, airspace stocking rate, and a trend toward higher atmospheric ammonia levels in the summer months. The trend to increased herd size has not been accompanied by the satisfactory control of pneumonia. It has been shown that pig-shed air polluted by alpha-hemolytic cocci and ammonia causes subclinical disease and production losses.34 Combination and Interaction of Environmental Risk Factors A computer-based guide can indicate how the prevalence of the disease can be influenced by the combined effect of risk factors. The expected prevalence is estimated by consideration of 11 risk factors that include the following: 1. Number of pigs in the same room 2/3. All-in, all-out versus continuous flow of pigs 4. Type of partitions separating adjacent pens 5/6. Presence or absence of diarrhea as a clinical problem 7/8. Liquid versus solid manure disposal 9. Ascarid control efficiency 10/11. Presence or absence of active Aujeszky’s disease. The temperature and humidity influence the penetration into the lungs of both primary and secondary pathogens by influencing the size of infected aerosol particles and the protective mechanism in the respiratory tract. Temperature and humidity also influence the sedimentation of infected particles in the air and the ventilation and stocking density. Pigs kept at high stocking densities and subjected to environmental temperature fluctuations, cold drafty conditions, and poor nutrition are more likely to suffer greater adverse effects from this disease.

Diseases of the Swine Respiratory Tract

VetBooks.ir

In a study of the effect of different housing and feeding systems considering liquid versus dry feeding in fully slatted and strawbased housing, there were no differences between in the lung lesions.35 Economic Losses and Importance In annual surveys completed by the American Association of Swine Practitioners, pneumonia consistently ranks as the most economically important disease in finishing pigs. The prime importance of enzootic pneumonia is in its economic effects on pig rearing. The disease adversely affects feed conversion efficiency and daily rate of gain under certain circumstances. However, the magnitude of these effects depends on the conditions in which the pigs are reared and has been a subject of much controversy. The complexity of pneumonia and its interactions with the environment make measuring the effect of pneumonia on performance very difficult. An accurate assessment of the biological and economic effects of enzootic pneumonia has been challenging because of the difficulty of conducting a controlled experiment in which pigs of equivalent genetic merit, both free of the disease and infected, are raised in an identical manner. In addition, studies on the association between performance parameters and the severity of lesions of the lungs have yielded widely variable results dependent on the management and environmental conditions and the different research design and techniques used. In general, there is a proportional relationship between severity of pneumonia and depression of performance but in other observations, this relationship was not found. Where pigs are raised under good management, infection of herds previously free of the disease has resulted in no adverse economic effect other than during the initial period of acute infection in the herd. However, in other situations adverse economic effects are associated with the disease. One study estimated a reduction of feed conversion efficiency as high as 22%, and although the effect of the disease is probably not this severe in most piggeries, a significant economic reduction can occur even under good management conditions. Because there is no universally accepted method of measuring the extent or prevalence of pneumonia in pigs at slaughter, the results of studies of correlations between the lesions and performance have been difficult to compare. In general, the economic loss associated with respiratory disease ranges from a 2% to 25% reduction in average daily gains. Some methods have been compared and the most informative procedure is to assess the percentage of lung involved and calculate a mean value for the herd sample. The relationship between the weight of pneumonic lesions from pigs at slaughter and their performance indicated that within a range between 3.32% and 74.5% for the

weight of a pneumonic lung, a 10% increase in the weight of pneumonic lung was associated with a decrease in mean daily gain of 31.4 g and a 13.2-day increase to slaughter at 104 kg live weight. There is a high correlation between rapid gross lung scores and detailed examination, which indicates that lungs can be visually scored accurately as they pass on a slaughter line. On average, mean daily gain decreases from 23 to 37 g for every 10% of the lung affected by pneumonia. However, the rapid subjective scoring of the lungs, adjusted for lung proportions, is considered adequate for estimating naturally occurring pneumonia and just as informative as detailed dissection of the lungs. Because the prevalence of pneumonia peaks at about 60 to 65 kg BW and then declines steadily to a very low level in pigs that are 125 kg or more, the age and weight at slaughter must be considered when evaluating the effects of the lesions on performance and when comparing results between different observations. Weight losses are more substantial in pigs affected early in life. In some studies, lung lesion scores detected at slaughter did not significantly correlate to growth indicators during any season. The gross lesions of mycoplasmal pneumonia heal over a 2-month period, which may explain why significant correlations are not found between growth indicators and lung lesions scores. The effects of the lesions on mean daily gains over an entire growth period may vary from one study to another because of the different times during growth when the lesions exerted their effects and in part to compensatory regrowth following recovery from the lesions. Radiographic examination of the lungs of pigs from 21 to 150 days of age, and gross examination of the lungs at slaughter revealed that lesions progress and regress dynamically throughout the life of the animals and examination at slaughter is an inadequate indicator of lifetime pneumonia.

CLINICAL SIGNS

There are very basically four signs of respiratory disease: • Sneezing is indicative of affliction of the nasal cavity gas, dust, or infection (PRCV, PCMV, or PAR). • Coughing is indicative of affliction of the larynx, trachea, and mainstem bronchi and upper bronchial tree because coughing is the only way to clear large amounts of infected debris (SIV, MH). • Dyspnea or difficult breathing is indicative of the terminal bronchioles and alveoli being affected (APP, PM, PRRS, and PCV2) • Parameters of growth may be affected when fever is involved or tissue damage is extensive, in which case the CNS (hypothalamus) instructs the systems to shut down

1073

so appetite, and therefore, growth, is reduced. Growth rate is reduced, daily gain falls, days to slaughter increases, and feed efficiency falls as growth is replaced by immunologic recovery. The principle sign of PRDC is pneumonia manifested as coughing, labored breathing, fever, lethargy, recumbency, anorexia, discoloration of the extremities/cyanosis, weight loss and slow growth, nasal and ocular discharges, and death. In small batches the disease may affect the group over a short period of time, and most may recover, leaving a few to become chronically affected, hospitalized, or having to be euthanized. In the larger batches with different age groups, there may be rolling waves of infection, or pneumonias may progress to pleurisy. Some pigs affected with the chronic form of mycoplasmosis may later develop acute pneumonia as a result of secondary invasion with Pasteurella or other organisms. A series of investigations has shown that PRRSV does not predispose to MH infection, although lesions are more severe in those pigs that both infections. MH does potentiate PRRSV induced disease and lesions. There may be an association between the seroconversion to PRRSV and the transmission of MH.

PATHOLOGY

Proliferative and necrotizing pneumonia (PNP) is a form of interstitial pneumonia that occurs in weaning and postweaning pigs. In an Italian study of 28 pigs PRRSV was found in 11 pigs, PCV2 in 4 pigs, and both viruses in the lungs of 8 pigs; in the other 5 pigs nothing was detected.36 A granulomatous lymphadenitis and pneumonia has been associated with Actinobacillus porcinotonsillarum in a slaughter pig.37 This organism was previously thought to be nonpathogenic. In the study in Denmark,3 no clear cut associations were found between pathogens and histologic lesions. They came to the conclusion that PRDC was more common than Mycoplasma pneumonia in Danish finishing pigs.

TREATMENT

There are many variables in an outbreak of PRDC, and it is essential to approach the problem in a sensible way. The first thing to do is to establish a diagnosis, probably using postmortem examinations and a variety of laboratory aids, such as IHC, ISH, and PCR. The definition of the primary pathogens from the secondaries and opportunists is the next major step. The third step is to treat the pigs quickly and effectively. The fourth step should be to assess the immune status of the herd and how to improve it. The fifth step is to understand the epidemiology of the agents in the herd and the health background of the herd. The last step is use the latest

1074

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

knowledge, strong biosecurity techniques, and modern vaccines correctly and to manage the units to use the best in management and environmental control.

MONITORING

Monitoring of respiratory disease has been achieved principally by slaughter checks. These involve snout inspection for the presence of progressive atrophic rhinitis. A cross section of the snout at the level of the 1st or 2nd premolar is examined. For the examination of the lungs, the percentage of the lung that is consolidated is calculated that is firm to the touch.18,38 Recently examination of digital images has been used.39 There is a significant negative association between pneumonia score and growth.40 In addition, the site of pleuritic lesions on the lungs can also be marked on cards and can be recorded as fresh or old fibrotic adhesions.

CONTROL

By definition PRDC is chronic respiratory disease (although there are periods of acute respiratory disease) in the continual production units of breeder/weaner and breeder/ feeder herds. In PRDC there are many potential agents, but there are some guiding principles that will help to maintain health. Maternal antibodies from sows on the same farm as the piglets will provide some protection, which wanes quickly. Young pigs then become susceptible, and if the numbers of pathogens are not too high they will develop active immunity without succumbing to disease. Infection follows the usual pattern of colonization, replication, excretion, and immune development. Disease may follow after replication and excretion, and the duration will depend on the level of replication and the agents involved. Older pigs are always a source of infection for younger pigs and will maintain a cycle of infection; therefore. they should be kept away from younger pigs, although sows should not be kept away entirely from young stock because their immunity is not then maintained. All-in, All-Out The first rule is all-in, all-out by age by building or by room. Complete disinfection after cleaning is then carried out, followed by drying and resting. The pig flow through the buildings must be established and maintained. Buildings • Make the production to suit the building provision, identifying bottlenecks. • Where necessary, alter the buildings (new divisions, new buildings, etc.). • Consider what the correct stocking rates are for the buildings.

• Ensure buildings are adequately ventilated to remove polluted air and excess heat without draughts or overventilation. Production Review productivity and consider batch production (i.e., a larger number of pigs less often) to enable filling and emptying of buildings. It is to effective to artificially construct a batch of different ages and hospitalized pigs. Evenness of production from the breeding units will prevent overstocking or understocking. Sick Pigs The sick pig is a welfare problem and a hazard in itself, so always hospitalize a sick pig as early as possible, treat, and cull it if no response. The hospital area should be well away from other pigs, and recovered pigs should not be returned to the mainstream. Diagnosis On farm or laboratory postmortem examinations should be used to achieve diagnosis if there are sudden acute cases. Cross-sectional blood sampling of the herd to establish epidemiologic patterns of pathogens is sometimes necessary. The use of slaughter pig information from the abattoir will indicate patterns of pathology. Active Control Lack of sound management cannot be compensated by use of medication and vaccination, but these may help. Partial depopulation with medication in Denmark has been described.41 Protection of your unit by the imposition of effective biosecurity from without and within the unit can be extremely beneficial in limiting the ingress of pathogens. The use of bird-proofing and rodent control is becoming much more important and in many cases the repair of the buildings is more important than other factors because cleaning and disinfection are pointless if there are areas where the organic matter can collect. Prophylactic or metaphylactic medication will help if targeted at the correct bacterial agent in feed or water. Vaccination for PRRS, PCV2, Mycoplasma hyopneumoniae, Hemophilus parasuis, and A. pleuropneumoniae, Aujeszky’s disease, and S. suis will also help. The effect of vaccination for PCV2 in pigs suffering from PRDC has been described.42 Additional strategies for PRDC will include partial depopulation and full depopulation as discussed for MH. REFERENCES

1. Ross RF. Anim Hlth Res Rev. 2006;7:13. 2. Nicholson TL, et al. Infect Immun. 2009;77:2136. 3. Hansen MS, et al. J Comp Pathol. 2010;143:120. 4. Palzer A, et al. Vet Rec. 2008;162:267. 5. Opriessnig T, et al. Anim Hlth Res Rev. 2011;12:133. 6. Fablet C, et al. Res Vet Sci. 2012;91:627. 7. Taira O, et al. Vet Microbiol. 2009;139:347.

8. Atanasova K, et al. Vet J. 2011;188:210. 9. Brockmeier S, et al. Vet Microbiol. 2008;128:36. 10. Ellis JA, et al. Am J Vet Res. 2008;69:1608. 11. Loving CI, et al. Microb Pathog. 2010;49:237. 12. Maes D, et al. Proc Cong Int Pig Vet Soc. 2010;30. 13. Dorr PM, et al. J Am Vet Med Ass. 2007;230:244. 14. Lin JH, et al. Vet Microbiol. 2006;115:111. 15. Hoeltig D, et al. Proc Cong Int Pig Vet Soc. 2010;196. 16. Probst I, Hoeltig D. Proc Cong Int Pig Vet Soc. 2010;602. 17. Nathues H, et al. Vet Rec. 2010;166:194. 18. Thacker BJ, et al. Proc Cong Int Pig Vet Soc. 2010;144. 19. Capuccio JA, et al. Proc Cong Int Pig Vet Soc. 2010;587. 20. Kim S, et al. Proc Cong Int Pig Vet Soc. 2010;584. 21. Lange E, et al. J Gen Virol. 2009;90:2119. 22. Smith GJD, et al. Nature. 2009;459:1122. 23. Valheim M, et al. Proc Cong Int Pig Vet Soc. 2010;588. 24. Fraile L, et al. Vet J. 2010;184:326. 25. Meyns T, et al. Vet J. 2011;187:368. 26. Martinez J, et al. Vet J. 2009;179:240. 27. Sorensen JT, et al. Revue Sci Tech. 2006;25:493. 28. Fablet C, et al. Vet Microbiol. 2012;157:152. 29. Fablet C, et al. Prev Vet Med. 2012;104:271. 30. Banhazi T, et al. J Agric Saf Hlth. 2008;14:21. 31. Pavicic Z, et al. Acta Vet Brno. 2006;75:533. 32. Renandeau D. Trop Anim Hlth Prod. 2009;41:559. 33. Otake S, et al. Vet Microbiol. 2010;145:198. 34. Murphy T, et al. Vet Rec. 2012;doi:10.1136/ vr.100413. 35. Scott K, et al. Anim Welf. 2007;16:53. 36. Morandi F, et al. J Comp Pathol. 2010;142:74. 37. Ohba T, et al. J Comp Pathol. 2007;137:82. 38. Bollo JM, et al. Proc Cong Int Pig Vet Soc. 2010;205. 39. Baysinger A, et al. Proc Cong Int Pig Vet Soc. 2010;659. 40. Pagot E, et al. Rev Med Vet. 2007;158:253. 41. Szancer J. Pig J. 2008;61:1. 42. Fachinger V, et al. Vaccine. 2008;26:1488.

PORCINE CYTOMEGALIC VIRUS (INCLUSION-BODY RHINITIS, GENERALIZED CYTOMEGALIC INCLUSION-BODY DISEASE OF SWINE) Porcine cytomegalic virus rhinitis (formerly inclusion-body rhinitis), associated with a beta herpesvirus (family Herpesviridae), is an extremely common, but generally minor, disease in young pigs. It was first recognized in 1955. The virus is now called porcine herpesvirus-2. It is associated with the porcine respiratory disease complex-1.

ETIOLOGY

The virus (PCMV; SuHV-2) belongs to the subfamily of beta-herpesviruses of the family Herpesviridae. The virions exhibit typical morphology of herpesviruses. There are believed to be no serotypes or genotypes, although there is some antigenic variability. It has not yet been found as a problem in xenotransplantation although it does grow in human fibroblast cultures.1

EPIDEMIOLOGY

The virus is present in the upper respiratory tract of nearly all herds and pigs (in excess of 90%), and the major infection site is the

Diseases of the Swine Respiratory Tract

VetBooks.ir

conchal (turbinate) epithelium. It does not affect other species. SPF herds established by hysterectomy techniques are not necessarily exempt, and congenital transmission of the virus has been demonstrated. High excretion occurs predominantly in the 2- to 4-week period after infection. Transmission is via the respiratory route through direct contact and aerosol infection, possibly also via urine, and usually perinatally. When the virus first enters a susceptible herd, both transplacental and horizontal virus transmission takes place. Antibody responses start quickly, so there are often no clinical signs but widespread infection.

PATHOGENESIS

The virus invades epithelial cells, especially those of the nasal mucous glands, to produce destruction of acinar cells and metaplasia of the overlying epithelium, and the major clinical manifestation is that of upper respiratory disease. Following infection, the virus may become generalized. In older pigs, generalization is restricted to epithelial cells of other organ systems, especially those of the renal tubules, and is clinically inapparent. However, in very young pigs the virus also shows a predilection for reticuloendothelial cells, and generalization may result in further clinical abnormality. The virus also crosses the placenta, so it is possible for intrauterine infection to produce fetal death, along with runting after birth and very early pneumonia, rhinitis, and poor piglet weights at weaning. Congenitally infected animals excrete for life.

CLINICAL SIGNS

The incubation period is generally 10 to 21 days. Clinically, the disease affects piglets up to approximately 10 weeks of age, but the age at manifestation in any herd can depend on the method of husbandry. The disease usually occurs when the virus is introduced into the susceptible herd or if for some reason there is a huge increase in the number of susceptible pigs. A wide age-spectrum of involvement may be seen initially when the disease is introduced into the herd for the first time. In most herds the disease affects pigs in the late suckler and early weaner stage. It is at its most severe in pigs under 2 weeks of age. Sneezing is the most prominent sign and frequently occurs in paroxysms and following play fighting. There is a minor serous nasal discharge that rarely may be blood-stained and also sometimes muco-purulent, with a brown or black exudation around the eyes. There may be coughing. The clinical course varies approximately from 2 to 4 weeks. All pigs within the group are affected, but there is usually no mortality. Neonatal pigs may die without showing signs. Generalized cytomegalic inclusion-body disease may occur in pigs exposed to intrauterine infection and usually occurs as an outbreak involving several litters. The

syndrome is characterized by sudden death and anemia. There is often a history of scouring within the group within the first week of life, and affected pigs show skin pallor and often superficially appear plump and well developed as a result of edema, especially in the neck and forequarter regions. Death, resulting primarily from anemia, occurs during the week 2 to 3 of life, and mortality within the group may approach 50%. Petechial hemorrhages have been a feature of the experimentally produced disease in gnotobiotic pigs but do not necessarily occur in field outbreaks. A moderate anemia producing a check to growth, but without significant mortality, which may be seen in recently weaned pigs experiencing the disease. Many survivors may be stunted. More serious effects from generalized infection are seen when piglets are exposed to heavy infection at a very young age. It also occurs when there are new imports and when intercurrent disease and poor nutrition reduce resistance. This commonly occurs in large herds with high-density continual throughput farrowing and weaning houses. In addition to upper respiratory disease, infection at this age may result in enteric disease, sudden death, anemia, and wasting, with a marked unevenness of growth within the litters. There may be complete blockage of the nasal passages. It is believed that the olfactory epithelium may be damaged so that there is no sense of smell and that piglets may not then eat, explaining the that so many die.

PATHOLOGY

demonstrated in exfoliated cells obtained via nasal swabs from live pigs. Small intranuclear inclusion bodies are found in the reticuloendothelial cells. These are best taken from several pigs at the height of clinical infection. Diagnosis by virus isolation is uncommon because the virus has proved difficult to grow, but it will establish in porcine lung macrophage cultures and immortalized cells. Antibody to infection may be detected by indirect immunofluorescent techniques. ELISAs have been developed to show both IgM and IgG responses. Recently a PCR has been developed and this showed that 59% of pigs tested positive. However, only 59% of PCR positive pigs had clinical signs and lesions consistent with inclusion-body rhinitis. The original experimentalists described the presence of intranuclear inclusions, cytomegaly, and karyomegaly as being pathognomonic. Virus isolation and PCRs can be used. The best PM samples are conchal mucosa, lungs, pulmonary macrophages collected by lavage, and the kidneys. PCMV can occasionally be demonstrated in the brain, liver, and bone marrow. Virus isolation is possible on primary or immortalized cells. Antibodies can be detected by IFA, which peaks at 6 weeks postinfection and remains quite high for 10 to 11 weeks. The development of serum antibody levels coincides with the disappearance of viremia.

DIFFERENTIAL DIAGNOSIS

Differential diagnosis includes CSF, enteroviruses, parvoviruses, PRRSv, PCV2, and PRV.

Gross changes are not seen often in pigs over 3 weeks of age. In pigs under 3 weeks it may be possible to see catarrhal rhinitis, hydrothorax, and edema in various tissues. In fetal infections there may be stillbirths, mummification, embryonic death, and infertility. Interstitial nephritis and random focal gliosis in the CNS with inclusion bodies can be additional findings, with petechiation in the choroid plexuses, cerebellum, and olfactory lobes. In the acute fatal syndrome most of the basophilic inclusions are seen in the capillary endothelium and sinusoidal cells of the lymphoid tissues. Multifocal hemorrhage and edema results from the vascular damage.

TREATMENT

DIAGNOSIS

SWINE INFLUENZA

Inclusion-body rhinitis is not a primary cause of atrophic rhinitis. However, it is probably contributory in lowering local resistance to infection and in predisposing to more severe infection with Bordetella bronchiseptica and other respiratory pathogens. The diagnosis of inclusion body rhinitis is commonly made following the demonstration of typical intranuclear inclusion bodies in histologic sections from electively slaughtered piglets. Large basophilic inclusion bodies are found in the mucous gland cells of the conchal mucosa and may also be

1075

There is no effective treatment, and none is warranted in most herds. With severe rhinitis, antibiotics may temporarily reduce the severity of secondary bacterial infection.

CONTROL

Control of severe disease rests with management procedures that avoid severe challenge to very young piglets. It is also possible to produce virus-free pigs from hysterotomyderived pigs, but it is necessary to monitor. REFERENCE

1. Whitteker JL, et al. Transplantation. 2008;86:155.

SYNOPSIS Etiology Influenza A virus subtypes H1N1, H1N2, and H3N2 of Orthomyxovirus. Epidemiology United States, England, Japan, Canada, Belgium. Worldwide. Young pigs. High morbidity, low mortality. During cold months. Antigenic diversity of virus. Aquatic birds are natural reservoirs. Spread between pigs, New strains develop,

Continued

1076

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Signs High incidence of anorexia, fever, thumps, muscle stiffness; recovery in several days. Clinical pathology Polymerase chain reaction (PCR) test to detect virus. Hemagglutination test and enzyme-linked immunosorbent assay (ELISA). Lesions Marked congestion of upper respiratory tract. Exudate in bronchi. Atelectasis. Suppurative bronchiolitis. Diagnostic confirmation Demonstrate virus in tissues. Differential diagnosis list: • Enzootic pneumonia • Hog cholera • Inclusion-body rhinitis • Atrophic rhinitis Treatment Antimicrobials for secondary infection. Control No effective measures available. Vaccines are in use in certain parts of the world.

INTRODUCTION Swine influenza is an important cause of broncho-interstitial pneumonia throughout all pig-keeping areas of the world. Real problems are associated with the changing viruses that cause the disease and the ability of rapid genetic change to occur by genetic drift or shift.

ETIOLOGY

Classical clinical swine influenza is associated with influenza A virus subtypes H1N1, H1N2, and H3N2 belonging to the Orthomyxovirus genus of the Orthomyxoviridae family. The three types occur together as in Korea.1 Other types have been isolated from pigs, but as yet have not established as widespread endemic strains. Only influenza A viruses are important in pigs. They occur in a large number of species, including humans, primates, pigs, horses, sea mammals, and birds. Avian viruses are more stable than mammalian viruses, where the rate of evolution is much greater. Specific subtypes vary in their ability to cross species barriers. Specific gene combinations do have a part to play in influenza virus species specificity.2 Unstable gene constellations in avian species become stable only in secondary hosts but may then adapt and circulate freely.3 The methods by which they cross the species barrier are not well understood and are probably polygenic.4,5 An isolate of a Korean H1N1 virus was very similar to a U.S. virus, suggesting that it had been transmitted possibly by birds.6 When new variants occur in pig husbandry they are usually found in the pig population before they acquire the ability to spread rapidly and become associated with disease. They are named using the following convention: A/species/localization/

isolate number/year of isolation, for example, A/Wisconsin/125/98. If no species is indicated, it is a human virus. They are described with reference to the hemagglutinin (HA or H) and the neuraminidase (NA or N) that project from the surface of the viral envelope. There are 16 HA and 9NA forms that can be distinguished antigenically and genetically, and all of these have occurred in waterfowl and shore birds. They provide a permanent source of infection, as does the water on which they float. The H binds to sialic acid and mediates the virus infection of the host and contains most of the antigenic sites. It is the viral receptor-binding protein and mediates fusion with the host-cell membrane. It is an alpha2-3-galactose linkage in avians and an alpha2-6-galactose linkage on the glycocalyx of epithelial cells in mammals.7 The HA and NA are associated with receptor binding and virus release.8 No combination of HA and NA has as yet been identified that will increase viral stability during interspecies transmission. The distribution of these receptors and the limited replication of avian viruses in swine complicate the picture.9 The pig possesses both types of receptor and has therefore been considered as a “mixing vessel” because it can be infected by both avian and mammalian viruses. The N protein catalyzes cleavage of sialic acid and thereby facilitates the virus cell entry by degradation of mucins. The NA and HA are also the main targets of the host immune responses. The segmented nature of the virus facilitates the changes in the virus. The surface HA and NA antigens undergo two types of change: antigenic drift and antigenic shift. Anitigenic drift involves small changes but the shift may involve whole segments of the genome being changed. If a cell is infected with two or more viruses, interchange of genetic material can take place. The 8 RNA segments encode for 10 or 11 proteins.10,11 It may take multiple mutations to make a distinct HA,12 and then this has to link with other gene segments compatibly to facilitate survival, replication, and transmission.13 For example, in the spread of the pandemic 2009, it appeared that the M segment was crucial to the transmission of the virus. It is possible that within the currently circulating strains, a reassortant will occur every 2 to 3 years.14 Three types are found worldwide H1N1, H3N2, and H1N2. In Europe three SIV subtypes are cocirculating: (a) an avian-like H1N1 that came from wild birds in 1979, (b) a humanlike H3N2 with HA and NA genes originating from human virus descendants of the Hong Kong/68 pandemic virus, and (c) a subtype H1N2 reassortant that acquired H1 from human influenza in the 1980s.

H1N1—CLASSICAL

In the United States, these were found on their own until 1998. They were very similar to the 1918 pandemic virus.15 Since the

appearance of other viruses, particularly the triple reassortants, there seems to have been an increase in the genetic diversity of the H1N1 strains in the United States (as also in the H1N2). A typical reassortant found in Ohio16 had genes from human (PB1), swine (NA, HA, NP, M and NS), and avian (PB2 and PA). Even though the viruses were isolated over only 3 years, there was evidence of antigenic drift.

H1N1—OTHERS

Humanlike H1N1 viruses have been found in Canada,17 and H1N1 viruses with the human H1 have spread across North America; these have commonly been isolated from swine disease outbreaks in the United States. A triple-reassortant H1N1 virus was found in China with the NP and NS genes from a classical swine influenza virus, PB1 from a human virus and HA, NA, M, PB2, and PA genes from an avian virus. Five genes were also closely related to H1N2 viruses founds in China (NS, NP, PB2, PB1, and PA).18

H1N1 AVIAN-LIKE

Wild bird H1N1 viruses were transmitted to pigs in the late 1970s and established a stable lineage displacing the classical H1N1 swine lineage; once this had happened, interspecies transmission was facilitated. An H1N1 isolated from a turkey farm in northern Germany in 2009 showed a high affinity with avian-like porcine H1N1 viruses circulating and suggested that turkeys may be a possible bridge between avian and mammalian hosts.19 The predominance of avian-like swine genes in the Thai pig population has been described.20 An experimental transmission of avian-like swine H1N1 has been described, and the virus transmitted through naïve and vaccinated pigs without causing clinical signs.21

H1N1 HUMANLIKE VIRUSES IN PIGS

Humanlike viruses were reported in pigs in China before the pandemic,22 and it was concluded that pigs may act as reservoirs for older human H1N1 viruses.

H1N1-PANDEMIC 2009

There is no evidence that the 2009 pig H1N1 pandemic existed in pigs before May 200923 and before it was reported in humans. Soon after its discovery in Canada in 2009,24 it spread rapidly around the world, and most pig cases are believed to originate from humans, although there was often no real proof until 2011.25 It has been shown that the virus is fully capable of causing a global problem for swine.26 The initial incursions of this virus into European pigs has been described27 from separate nonlinked sites, suggesting infection of pigs from humans. The global spread from an animal source has

Diseases of the Swine Respiratory Tract

VetBooks.ir

been described.28 It has established itself in pig populations in face of relatively high levels of herd immunity to other viruses. In the Norwegian pig population, there was no prevalence of influenza until the infection of pigs from humans with (H1N1) pdm09.29 The virus is a reassortant of genes from the most recent triple reassortant in North America and the European avian-like subtype H1N1 viruses.30 The precursors of this virus may have existed in swine for a long time, which suggests that the evolution has occurred over a long period.30,31 A direct precursor has not been recognized.32,33 The situation was summarized.34 It transmits very effectively between pigs.35 The evolutionary characteristics of the H1 gene of the pdm2009 virus are different from the seasonal human viruses and the swine H1N1 viruses.36 The pandemic virus seems to cocirculate and interact more intensely with the endemic SIVs lineages and gives rise, it seems, to more reassortants, the properties of which have yet to be seen.37 A mono-reassortant of the NA from an H1N1 with the pandemic occurred in Hungary.38 In a study in Germany, the N2 was from three different porcine lineages in an H1N1pdm backbone. Six new strains of the pdm-like (H1N1) 2009 strain of H1N1 were isolated and characterized in Poland. They belong to one lineage.39 The pigs in finishing and growing sectors experienced acute onset of respiratory signs. There was anorexia, poor conception rates (50% lower), high morbidity (up to 100%), and low mortality at 2% to 3% in growers and 1% to 2% in finishers. At postmortem there were depressed, welldemarcated. pale purple areas of consolidation in all lobes. Novel reassortants have followed with this 2009 virus, and it was pointed out in 2010 that although the virus may be of swine origin. significant viral evolution may still be ongoing40 and others starting with a 2010 virus in Hong Kong.41 In this virus, only the NA gene of the 2009 pandemic was reassorted. A novel swine reassortant has been described in the United Kingdom with all the internal genes from the 2009 virus and HA and NA genes from a swine subtype H1N2 virus.42 It is not clear if this virus can be transmitted between pigs. A novel reassortant has been found in Canada from ab H3N2 and a pandemic (H1N1) 2009 virus on several pig farms and also in mink.43 In another reassortant, the NA glycoprotein of the pdm09 virus has been replaced by the NA gene from either H1N2 or H3N2 European swine viruses.44 Other reassortants of the 2009 virus have been discovered since from a variety of countries Italy, Argentina, Germany, China, Thailand, and the United States.44-50 Nine reassortants have been described across the United States.51

A reassortant of the pdmH1N1 2009 virus with an H3N2 virus from healthy pigs has been reported in Thailand.52

H1N2

Since 2005, the human HA gene in H1N2 has spread across North America. A novel reassortant in H1N2 had the NA and HA from the recent H1N2 isolates in the United States and four internal genes (PB2, PB1, PA, and NS) from the contemporary swine triple reassortants in circulating strains, known as the TRIG, but the NP and the M genes were derived from the 2009 pandemic H1N1.53 An avian-like H1N2 SIV generated by reassortment of circulating avian-like H1N1 and H3N2 subtypes in Denmark has been described.54 The Danish H1N2 has an avianlike H1 and differs from most other H1N2s in Europe and North America. These have H1 genes of human or classical swine origin, respectively. The variant is also circulating in Italy and Sweden. The infection dynamics are similar to the those of the assorted H1N2s and similar to the older avian-like H1N1 subtype. A novel reassortant influenza A (H1N2) virus derived from A (H1N1) virus Japanpdm09 has been described for the first time in Japan.219

H3N2—CLASSICAL

H3N2 variants arrived in the United States from 1998 onward (North Carolina, Iowa, Minnesota, Texas), although they may have circulated previously and had been unable to establish a stable lineage. Most are triple reassortants from human (HA, NA, and PB1), swine (NS, NP, and M), and avian (PB2 and PA) lineages. By 1999 these were widespread in the United States, and a double assortant that had also been found had not spread widely. These are capable of being placed in one of three phylogenetically distinct humanlike lineages (clusters). The third cluster seems to be dominant and some have developed into a fourth cluster.55 A study of 97 isolates showed that genetic and serologic differences existed between North American isolates56 and that they show tendencies to reassortment. Once established, these have spread rapidly and evolved.57

H3N2—NOVEL

A noncontemporary H3N2 virus was found to be a wholly human H3N2 virus.58 Triplereassortant H3N2 SIVs were isolated from pigs59 and have formed a stable lineage in Canadian swine. Novel H3N2 viruses in the United States in humans have been linked visits to state fairs and contact with pigs. Similar occurrences have been found in the past, but these have not had a component of H1N1 in the virus, as does this 2011 variant. Seven novel H3N2 viruses were isolated from U.S. pigs between winter 2010 and

1077

spring 2011 containing internal gene segments from the pandemic H1N1 2009.60 The evolution of novel H3N2 viruses in North American swine has been described.61 A novel avian-like H3N2 containing an H5N1 highly pathogenic segment has been described in southern China.62 An influenza A (H3N2) virus from pigs was isolated from pigs and its biological properties reported.63 The virus produced mild interstitial pneumonia with marked oronasal shedding for about 14 days. Because there is likely to be little cross immunity to these strains, they may cause disease in both humans and pigs in the future.

OTHER VIRUSES

Two H5N1 influenza viruses have been isolated from swine in Jiangsu Province in China, and the authors have suggested that swine are naturally infected with H5N1 virus.220 This was similar to the situation in Indonesia.64 Quite often these reach pigs from avians, particularly ducks, including H1N1, H3N2, H3N3, H4N6, H5N1,64-66 and H9N2. H2N3 viruses were isolated from farms in central United States67,68 and were probably of waterfowl origin. The ability of this virus to live in three different mammalian hosts suggests that it is well adapted. An H3N1 SIV has been isolated from pigs with respiratory disease in Korea69 and also in Italy,70 where the HA has been acquired from a human virus, and the other genes came from the currently circulating viruses in the swine population. Novel viruses can occur in pigs at any point in time. An avian H4N6 virus appeared in Canada in 1999 and was associated with a lake on which there were large numbers of waterfowl. Ducks shed large amounts of virus, and this can be recovered from lake water. An avian-like H4N8 SIV was discovered in southern China.62 An assessment of the reassortant rates of the European strains of SIV suggested that there was one reassortment every 2 to 3 years, and we should expect these to occur in the future between the swine strains and the new human pandemic strain (2009).14 A high level of genetic compatibility between swine-origin H1N1 and highly pathogenic avian H5N1 influenza virus was shown.71 The avian H5N1 viruses in birds in Indonesia have been transmitted to pigs on numerous occasions72 but appear to become attenuated. H5N2 reassortant viruses have been characterized from pigs in Korea.73 A serologic surveillance of H1N1 viruses in China showed that there was no naturally occurring H5N1 infection in pigs.74 A highly pathogenic turkey H5N1 virus failed to infect pigs cohoused with infected chicks or chickens.75

1078

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

A H6N6 virus was found in swine in China and seems to have adapted from domestic ducks.76 The isolation and characterization of two H5N1 influenza viruses from swine in Jiangsu Province in China has been described.220 The H5N1 virus has spread to a range of avian and mammalian species but has not been fully characterized in the pig. Both swine viruses bound preferentially to avian-type receptors. The findings suggest that pigs are naturally infected with avian H5N1 viruses and are a potential zoonotic threat. In a study of enhanced infectivity of H5N1 highly pathogenic avian influenza virus in pig ex vivo respiratory tract organ cultures following adaptation by in vitro passage.221 It was suggested that the mutations in the H5N1 virus may provide a replication or infection advantage in pigs in vivo and that pigs may continue to play an important part in the ecology of influenza viruses, including those of avian origin. An H7N2 virus was isolated in South Korea and was a recombinant from an avian H7N2 and H5N3 virus.77 H9N2 SIVs have been described in China,78 where the six internal genes are from H5 viruses and the HA and NA from the H9 lineages. In a survey in China, 54 genotypes were identified including 19 novel genotypes,79 and there is a continuing evolution of these viruses. In this study, at least five antigenic groups were recognized, and during the period of 2002 to 2003 there was a considerable antigenic drift. Human H7N9 IV replicates in swine respiratory tissue explants.80 Three Chinese isolates all replicated in tracheal and bronchial explants. These viruses were originally avian viruses that appeared in humans in China with over 130 cases, with a mortality of 32%. The surface proteins are probably from ducks and the internal genes possibly from chickens. There are two lineages reported at the moment. Collectively these viruses could lead to another pandemic. The infectivity, transmission, and pathology of these viruses in pigs has been described.81 An H10N5 virus has been isolated from pigs in central China.82 There is no evidence as yet that the “bird” viruses H10N8 and H7N9 poultry viruses that have killed people in China are as yet occurring in pigs in China.

EPIDEMIOLOGY

The segmented nature of the viral genome is a critical structural feature that enables the viruses to be reassorted. Since 1998, H, N, and PB1 polymerase genes from human viruses; M, NS, and NP genes from classical swine viruses; and PA and PB2 polymerase genes from avian viruses have also been found. Occurrence Influenza viruses are ubiquitous in pigs worldwide with the exception of Norway until the 2009 pandemic.83

A seroprevalence and genetic characterization of five subtypes of influenza A viruses (H1, H3, H5, H7, and H9) in the Chinese pig population has been described.84 H1 is the most common, followed by H3. A study in the United Kingdom suggested that at least 52% of farms had antibodies to at least one type.85 A Belgian study involving seven European countries86 showed all had antibodies, but the Czech Republic, Ireland, and Poland had relatively lower levels. Both H1N1 and H3N2 are found in Poland but at quite low levels.87 Chinese studies suggested that there was 31.1% positive to H1 and 28.6% positive for H388. In a recent study in southern China, over 50% of the pigs tested had a HI titer to one or more influenza H1N1 viruses, and most commonly pdm/09-like viruses. One group had Eurasian avian-like swine H1N1 surface genes and pdm/09 internal genes.50 The viruses were similarly widespread in Korea88,89 and also in Malaysia.90 In Canada, 83.1% of the sows and 40.3% of the finishing pigs were positive for H1N191 but less than 10% to the Colorado and Texas strains of H3N293. In Argentina, over 70% were positive for H1N1 and H3N294. In Brazil, 46% were positive for H1N1.93 Swine influenza first appeared in the United States immediately following the 1918 pandemic of human influenza (Spanish flu), and it was generally believed that it was caused by adaptation of the human influenza virus to swine. Nucleotide sequencing of the genes coding for the internal virus proteins indicates that the human pandemic H1N1 strain and the classic swine strain H1N1 have a common avian ancestor. It is suggested that a virulent avian strain H1N1 entered the human population in 1918, causing the pandemic. The pandemic virus was then introduced into the swine population, where it has persisted unchanged. In contrast, this classical swine influenza was seen in the United Kingdom in 1941 but then disappeared until it was seen in Czechoslovakia in 1950 and Germany in 1959. Influenza was not seen again until observed in swine in Europe in 1979, possibly following importation of pigs from North America, associated with a virus antigenically related to contemporary avian H1N1 strains found in ducks. These avian-like strains have been the most common since 1979. Swine influenza still occurs in the United States, and viruses of the H1N1 lineage were the dominant cause of SIV from 1930 to the 1990s. These were highly conserved (relatively unchanged), but new antigenic and genetic variants did occur. Classical H1N1 viruses have also been isolated from pigs from South America, Europe, and Asia. Wild pigs also have H1N1. In the 1980s there were many genetic mixings between avian-like H1N1 and human-like H3N2 viruses. In

1992 many outbreaks of classical swine influenza occurred in England, associated with a group of H1N1 viruses that were distinguishable from classical swine viruses, the European swine viruses, and human H1N1 viruses, all of which are known to be circulating in pigs. Influenza A virus subtypes H1N1 and H3N2 are endemic in pigs in Great Britain. Two distinct antigenic variants of H1N1 viruses have been associated with outbreaks of swine influenza, one of which was probably transmitted from birds to pigs in the early 1990s. The H1N2 subtypes isolated from pigs in Great Britain appear to have originated from a human H1N1 virus, which was circulating in the pig population in the 1980s, and from swine H3N2. It is suggested that the H1N1 viruses have disappeared from the human population, and the pig population provides a reservoir for the virus. Serologic surveys indicate that a swine H1N1 influenza virus has circulated in the swine population in North America for many years. Recent isolates from Quebec possess a hemagglutinin distinguishable from subtype H1N1. Transmission of viruses between pigs and humans and vice versa have shaped the current epidemiology of influenza viruses in North America. Epidemics of swine influenza have also occurred in Japan, Canada, Belgium, and France. In North America, human H3N2 have been found much less often than in the rest of the world, but the very recent introduction of H3N2 from humans to pigs was probably the major factor in the emergence of the recent strains. Mixtures of human and classical virus genes have been isolated from pigs in Asia and the United States. H3N2 viruses with human H and N genes and avian internal protein genes have been isolated from pigs in Asia. This type of H3N2 has been found in Korea and is currently the dominant H3N2 virus in pigs in Europe. Since 1998 double and triple reassortants have been isolated from pigs in the United States. The North Carolina virus had three human genes and five swine genes. They include human H and N genes, genes from swine H1N1 viruses, and two others from avians. All the reassortant viruses found in North America have the triple-assortant gene complex (avian PA, PB2; the NS, NP, and M genes of classical swine lineage; and the PB1 of human gene lineage). This suggests that this set of reassortants can more readily accept changes in NA and HA) Prior infection with swine influenza viruses is a barrier to infection with avian influenza viruses.94

SEASONALITY

A study of circulating viruses in five European countries showed that isolation of viruses was possible throughout the year, especially during winter and spring.95

Diseases of the Swine Respiratory Tract

VetBooks.ir

Soon after the occurrence of the H3N2 viruses, new H1N2 viruses arose in the United States, where the human H3 had been replaced by a porcine H1 and then spread. They had been known elsewhere in the world for some time: Japan, France, Germany, and Taiwan. They were described in the United Kingdom, where they were found to be the most severe cause of pathology associated with the SIV viruses. These were all reassortants between human H3N2 and classical H1N1. Human H3N2 and avian H1N1 were isolated in the United Kingdom and were then found to have spread to Europe. They are usually human H and N and the rest avian genes, but one Italian virus has an avian H1. They have shown considerable genetic drift in Europe. Subtype H3N2 has been isolated in Canada from pigs with severe proliferative and necrotizing pneumonia (PNP), although this PNP is probably associated with PRRS and PCV2. Serologic surveys indicate the infection is widespread in the swine populations in some countries. The first unusual virus to be found in pigs was an H9N2 introduced to pigs in South East Asia, probably from land-based poultry. Further problems occurred in the autumn of 1999 when an avian H4N6 was found in pigs with pneumonia on a commercial swine farm in Canada. Since then the avian H5N1 has appeared in pigs in China and is being carried west by bird migrations into Russia. The potential of avian viruses to spread to pigs and persist in pigs is unknown. Even if the viruses do not replicate, they can contribute viral genes to other pig viruses. This is the reason for continual surveillance of SIV viruses. These were wholly avian viruses that were of North American lineage. It was the first report of an interspecies transmission of an avian H4 virus to domestic pigs under natural conditions. The disease usually affects young pigs, but all ages may be affected. Typically, sudden-onset epidemics occur with a high morbidity rate but with a low case-fatality rate of less than 5%. Loss of body condition is marked, which is usually the important cause of financial loss, although occasionally death losses may be extensive if the pigs are kept under inadequate conditions or if secondary bacterial infections occur. Abortions and deaths of newborn pigs have also been reported as causes of loss in this disease. A low level of infection was reported in Poland in 2007 in pigs, wild boar, and animal keepers.96 The 2009 pandemic first affected pigs in Canada97 and has since been found worldwide: Norway,98 Italy,99 Canada,100,104 Argentina,101 South Korea,102 Thailand,103 and Europe.27

Risk Factors Animal Risk Factors In a study in the Netherlands, it was shown that at the end of the finishing period, the seroprevalences in farrow to finish herds and specialized finishing herds were 44.3% and 62.0% for H1N1, 6.6% and 19.3% for H3N2, and 57.2% and 25.6% for H1N2. The incidence for all three types was highest at the beginning of finishing in farrow to finish and at the end in finishing herds.105 Risk factors include high pig density, large herd size, high replacement rates, and purchase of pigs.90,91,106,107 Young, growing pigs are most susceptible. The viral infection is commonly complicated by bacterial infection caused by Haemophilus parasuis, A. pleuropneumoniae, and possibly other opportunists of the upper respiratory tract of the pig. When an epidemic occurs, most of the pigs in the herd are affected within a few days, which suggests that all animals are previously infected and that some risk factor, such as inclement weather, precipitates the epidemic. The agent also contributes to the PRDCx. In a study in Korea, 14 of 105 cases had SIV, whereas in Iowa it has been reported in 19% of the cases of PRDC. Environmental Risk Factors Epidemics occur mainly during the cold months of the year, commencing in the late autumn or early winter and terminating with a few outbreaks in early spring. Several days of inclement weather often precede an outbreak. Three risk factors for SIV were identified on a survey of Belgian finishing farms, where H1N1 was found in 71% and H3N2 was found in 22%. There was a close association between H1N1 and H3N2. H1N1 appeared to be associated with fully slatted floors, increasing numbers of pigs in the locality, and dry feeding. H3N2 was associated with the purchase of pigs from more than two herds, increasing numbers of pigs locally, and natural ventilation. Pathogen Factors It has been shown that prior infection with swine influenza viruses in pigs is a barrier to subsequent infection with avian influenza viruses.94 Molecular microbiology has now revealed the antigenic diversity of the virus. Several different H and N antigens have been identified and grouped on the basis of serologic tests, which refine the diagnosis and reveal more about the epidemiologic relationships. The H3N2 strain similar to H3N2 strains found in the human population has been isolated from an outbreak in England. Two antigenically distinct H1N1 influenza A viruses were isolated during an outbreak of respiratory disease in swine in Canada in 1990 to 1991. One is a variant of the swine H1N1 influenza virus that is widespread in the American Midwest, whereas

1079

the other is similar to the virus isolated from swine in 1930. This suggests that influenza viruses can be maintained for long periods in swine herds, especially in certain geographic areas. It is proposed that the antigenic diversity of these viruses may be attributable to the result of drifts in the population of circulating swine influenza viruses in an area.7 The antigenic diversity oligonucleotide analysis of strains isolated from outbreaks in Sweden indicated a similarity with the Danish strain. One of the Swedish strains was closely related to the U.S. strain. The H1N1 strain of the virus can be found in pig tissues at slaughter but it does not persist for more than 2 to 3 weeks in deep frozen or refrigerated storage. Virus circulation in weaned pigs may maintain infections in herds,108 and the introduction of susceptible pigs at regular intervals will maintain this circulation. Methods of Transmission Of most importance is that in birds, influenza viruses mainly affect the intestinal tract (without clinical effects), but in mammals, replication occurs mainly in the respiratory tract (with illness). The right combination of NA and M genes is necessary for the replication and transmissibility of influenza virus infections in pigs.109 The natural reservoir of influenza A virus is aquatic birds. Various subtypes have been established in other species, such as influenza A H1N1 viruses, which infect human and different animal species. The influenza viruses may be transmissible between humans and pigs. Swine are the sole animals known to be susceptible to influenza A viruses of human, swine, and avian origin. Swine may become infected with related type A human influenza strains during epidemics of human influenza, but they show no clinical signs of infection. The human strains have been isolated from pigs in Hong Kong, and pigs may serve as a reservoir for pandemics in humans and a source of genetic information for recombination between human and porcine strains. In Japan, pigs may be seropositive to the H1N1 human viruses relative to human H1N1 influenza epidemics and seropositive to human H3N2 viruses unassociated with human epidemics of disease. In Czechoslovakia, influenza A viruses are brought into pig herds by carrier people. Pigs can be naturally infected with a range of avian influenza viruses. There have been at least three independent introductions of distinct wholly avian viruses into pigs. The virus in the late 1970s spread throughout Europe and the United Kingdom and became a major cause of SI. These viruses also undergo drift. Elsewhere in the world antibodies against H4, H5, and H9 viruses have been isolated from Asian pigs and avian H4N6, H3N3, and

1080

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

H1N1 viruses have been recovered from pigs in Canada. Aerosol transmission is more efficient at low temperatures and low humidity because the virus is more stable under these conditions.110 Aerosol transmission of a novel swine origin H1n1 virus was shown in China.111 In water the avian viruses survive better at low temperature and salinity and high pH.112,113 The avian virus survives better on nonporous surfaces rather than porous ones114 and, if there is mucus, much longer.115 Swine are susceptible to both human and avian viruses because they have receptors on their respiratory epithelial cells for both avian (receptor SA 2, 3 Gal) and human (receptor SA α 2, 6 Gal). Several reassortants have been isolated from pigs in the United States and other parts of the world. Thus swine have an important role in the ecology of influenza A viruses and are regarded as a “mixing vessel” for the introduction of reassorted viruses into the human population. There is a report claiming that outbreaks of influenza in turkeys followed outbreaks of swine influenza in pigs from nearby swine herds. Swine and other influenza viruses have also been isolated from cattle, and experimental inoculation of calves has been successful. The swine influenza virus may cause natural infection in cattle and the virus can be transferred to uninoculated calves. The primary route of infection is through pig-to-pig contact116,117 via the nasopharyngeal route. Peak shedding occurs 2 to 5 days postinfection (>107 infectious particles/mL at a peak) but also by aerosols and contaminated fomites.118 The rapid spread of infection from pig to pig occurs by inhalation of infective droplets. The disease may appear almost simultaneously in several herds within an area following the first cold period in late autumn. The virus can persist in infected swine, which can act as convalescent carriers and be the reservoir of the virus between epidemics. However, the experimental inoculation of a swine influenza virus into specific-pathogen-free (SPF) pigs resulted in a mild disease and the period of viral shedding was shorter than 4 weeks. Water contaminated with bird droppings has been implicated as a source of influenza virus in several swine outbreaks.68 Fomites and aerosols92 are probably important in the transmission of influenza.119-122 Insects may be important (certainly in avian influenza123) and blowflies have been implicated.124,125 Long-distance pig travel via transport may help spread.126 International trade may also facilitate the intercontinental spread of viruses.127 Immunity An infection with live virus also stimulates mucosal immunity and cellular immunity,

whereas inactivated vaccines only stimulate a limited serologic (HI) response. Preexisting immunity in European pigs to established SIV strains may partially protect against (H1N1) 2009 virus, but the extent of such protection needs to be assessed.128 Many of the host defense cells have sensors that ultimately up-regulate the production of interferons, up-regulate other cells, and activate them through cytokines and in general increase the production of host antiviral proteins. The flu virus survives in part by blocking the release of interferons. Both cell-mediated immunity and humoral responses are important. A high HI titer provides better protection against challenge than a low HI titer. The levels of IgA seem to be more important in providing some protection against heterologous viral strains. It is the antibody-mediated immune reactions at the mucosal level, not the systemic level, that are important in protecting the respiratory tract. Improved adjuvants may aid the efficacy of inactivated vaccines. They do not prevent infection, but they can mediate the killing of infected cells. The immune response is rapid and completes elimination of the virus within 1 week. Antibodies decline by 8 to 10 weeks. The IgA levels in nasal washes are the most important defense. There is limited cross-protection between different viruses, and protection after vaccination is more virus specific. Maternal antibody rarely prevents infection with influenza viruses and only provides partial protection. Maternal protection will last from 4 to 14 weeks, with no pigs being completely protected from nasal virus shedding upon challenge, but at least the lung is protected. Pigs with a high maternal antibody level did not develop an immune response. It was reported that there was enhancement of pneumonia by inactivated vaccine used in the face of an H1N1 challenge.129 Maternal antibody does not cross protect between subtypes. Pigs infected or vaccinated with European SIVs frequently have cross-reactive antibodies to pandemic (H1N1) 2009 virus and related North American SIVs. Prior infection with an H1N1 SIV partially protects pigs against a low-pathogenic H5N1 avian influenza virus.130

ZOONOTIC IMPLICATIONS

Only influenza A viruses are zoonotic. The suspected cases were reviewed.131 It is highly likely that in the future, further viruses will emerge from animal species to infect humans and vice versa. People who work with pigs are at an increased risk of zoonotic influenza virus infection132 (including farmers, meat processing workers, and veterinarians)133,134 and should be vaccinated.135 The Ohio outbreak of H1N1 at a state fair is an example. H2 viruses have been absent from the human population since 1968 and as such

will present a huge problem if they suddenly turn up as a zoonosis. However, an H2N3 infection in pigs was not transmitted to humans from ill pigs.136 In the United States, there were only 11 reported zoonotic cases between 2005 and 2009.137 The human pandemic 2009 H1N1 virus has its closest relatives in strains of H1N1 in swine from North America and occasionally from turkeys. There are probably at least two swine ancestors for this 2009 pandemic. Subclinical infections at Ohio fairs from 2009 to 2011 were described.138 The influenza A virus (OH07) isolated from humans that attended an Ohio state fair is pathogenic in pigs and fails to cross-react with many swine H1 antisera. The virus gene segments were similar to those circulating in swine viruses, although there were numerous nucleotide changes leading to differences in amino acid composition.139 Swine influenzas pose a significant health risk to humans ever since the first human and porcine outbreaks in the United States in 1918. By 1970, there was evidence that people who came into contact with pigs through their jobs became infected with the viruses, and a virus was isolated from pigs and workers. There is very little evidence of maintenance of human H1N1 in the pig populations, but human H3N2 strains have been recovered regularly from pigs in Asia and Europe. The drift that has taken place in pigs of former human H3N2 has also been minimal compared with the rate of drift in the human population. The viruses from pigs found in humans have been reviewed. Poultry and swine workers should be vaccinated in swine pandemic planning.135

PATHOGENESIS

Classical swine influenza was originally described as a disease of the upper respiratory tract, the trachea and bronchi being particularly affected, with secondary bacterial pneumonia as a result of Pasteurella multocida. However, recent descriptions of the lesions in naturally occurring cases and in the experimental disease indicate that the primary lesion is a viral interstitial pneumonia. Viral replication takes place in the epithelial cells of the nasal mucosa, tonsils, trachea, lungs, and tracheo-bronchial lymph nodes. No other sites have been detected, and viremia is of low titer. Inoculation of the H1N1 strain of influenza virus isolated in England from pigs with clinical disease into 6-week-old pigs caused fever, coughing, sneezing, and anorexia. A widespread interstitial pneumonia, with lesions in the bronchi and bronchioles, and hemorrhagic lymph nodes were characteristic. The H3N2 swine influenza virus isolated in Canada is associated with a proliferative and necrotizing pneumonia (PNP) of pigs, and there is evidence the strain may be related to A/Sw/ Hong Kong/76H3N2 swine influenza virus.

Diseases of the Swine Respiratory Tract

VetBooks.ir

There is recent evidence that this PNP is more a feature of PRRS and PCV2 than SIV. A new antigenic variant of H1N1 swine influenza A virus isolated in Quebec has been associated with proliferative and necrotizing pneumonia of pigs. In the United Kingdom, there has also been recorded an H1N7 that included both equine and human influenza genes. It was of low pathogenicity for pigs, found on only one farm, and did not establish in the pig world. Reassortant H3N1 viruses from human and classical swine H1N1 have also been seen in the United Kingdom and also in Taiwan. The virus causes an acute infection with shedding beginning on day 1 and finishing by day 7. Infected cells in the respiratory tract are reduced by 2 to 3 days postinoculation. Most of the effects of the infection are caused by the production of proinflammatory cytokines (IFN-α, TNF-α, IL-1, and IL-6.). Pigs have receptors for both avian (sialic acid–alpha-2,3 terminal saccharides (SAalpha-2,3) and mammalian viruses (SAalpha-2,6) in the upper respiratory tract. Both types have been detected in major porcine organs.140,141 In experimental infections, SIV was widely distributed in bronchi, but it was also present in epithelial cells of the nose, trachea, bronchioles, and alveolar type I and II cells in severely affected animals. The avian virus was found in the lower respiratory tract, especially in alveolar type II cells and occasionally in bronchiolar epithelial cells. Receptor 2,6 was the predominant receptor in all levels of the tract, but the 2,3 was found only in small numbers in the bronchioles and in the alveoli. The receptor expression of both types of receptors was reduced in influenza-affected areas compared with nonaffected areas.142 The distribution of receptors is similar in the pig to that of humans, and as in humans, avian viruses prefer to infect the alveolar cells. The in vitro attachment of virus to the upper and lower respiratory tract tracts of pigs has been characterized.143 The pathogenicity of SIV lies in its ability to elude host antiviral immune responses. In pigs SIV infection induced long-lived increase of CD8+ T cells and local lymphoproliferative responses.144 The activation of cell-mediated immunity or cytotoxic Tlymphocytes depends on the efficient delivery of signals by antigen presenting cells. Dendritic cells are the most potent APCs. A study on porcine dendritic cells (DCs) has recently been published.145 In one study,146 it was shown that DCs could infect susceptible cells by close contact. The swine, human, or avian viruses differentially activate porcine dendritic cell cytokine profiles.147 There is an important role for IFN-α (induces fever and a transient rise in neutrophil counts) with IL-6 and IL-12 induction and an important role of these three

cytokines in the symptoms of swine influenza.148 There is a strong up-regulation of additional cytokines (IFN-α and IL-12) and several acute-phase proteins during the acute stages of a swine influenza virus infection. These produce inflammation, fever, malaise, and loss of appetite. The depth of infection in the lung probably determines how much of these cytokines are produced. Contrary to widespread belief, there is no evidence that the virus causes reproductive failure in swine. The experimental inoculation of seronegative pregnant gilts did not reveal any evidence of transplacental transmission of the virus to the fetus. The pandemic H1N1 influenza virus causes disease in pigs and up-regulates genes related to inflammatory and immune responses. The virus is effectively shed from the nasal passages. Pigs infected with the pandemic virus mounted an early potent immune response, and it has been shown that such a response is associated with an increased viral pathogenesis. It also produced a higher proinflammatory cytokine response when given to macaques.149 The PB1-F2, which is expressed from a +1 reading frame of the viral RNA polymerase subunit PB1, is able to induce apoptosis and promote inflammation.150 Dysregulation of lipid metabolism also occurs at the site of primary infection.151 The pandemic 2009(H1N1) virus was shown to be more pathogenic in ferrets than the standard seasonal H1N1 virus with more extensive viral replication taking place in trachea, bronchi, and bronchioles and the more normal nasal cavity.152 The virus replicates to higher titers in the lung tissues. It showed less efficient respiratory droplet transmission in ferrets.153 In patients with pandemic A (H1N1) pdm09, it was found that the numbers of dendritic cells and T cells were significantly reduced compared with controls. On the other hand, the frequency of natural killer cells and T-regulatory cells increased. The concentrations of plasma interferon (IFNα/γ) and interleukin (IL-15) were significantly higher than in the control group.154

CLINICAL FINDINGS

The patterns of disease in farms may vary considerably from an endemic form, with waves of infection to single epidemic outbreaks depending on the strains of virus involved.155 It is essentially a herd disease. The signs have not changed over the 80 years. After an incubation period of 1 to 7 days (usually 1-3), the disease appears suddenly, with a high proportion of the herd showing fever (up to 41.5° C [107° F]), anorexia, and severe prostration. The animal is disinclined to move or rise because of muscle stiffness and pain. Labored, jerky breathing (“thumps”) is accompanied by sneezing and a deep, painful cough that often occurs in paroxysms. There

1081

is congestion of the conjunctivae with a watery ocular and nasal discharge. Sometimes there is open-mouth breathing and dyspnea, especially if the pigs are forced to move. Morbidity is usually 100%, but mortality is rarely above 1%. In general, the severity of the illness appears greater than it truly is, and after a course of 4 to 6 days, signs disappear rapidly, depending, in part, on the level of colostral antibody. However, there is much loss of weight, which is slowly regained. Clonic convulsions are common in the terminal stages in fatal cases. The condition may continue to affect the herd for several weeks as the disease spreads, especially so if the herd is outdoors and the population dispersed. The new H3N2 reassortants in the United States have been associated with respiratory disease but also spontaneous abortion in sows and death of adult pigs. The clinical signs are dependent on immune status but are also influenced by age, infection pressure, concurrent infections, climatic conditions, housing, and, most of all, by the secondary infections. particularly bacteria. The clinical and epidemiologic characteristics of pdmH1N1 2009 virus in pigs have been described.156 There are differences in disease presentation, spread, and duration of infection. These factors include whether they were outdoors or housed, age of the pigs, intercurrent disease, and management. In breeding pigs the infection was mild or inapparent, with a more typical clinical appearance detected in their progeny. Mortality was low unless complicated by other diseases, especially S. suis infections. The virus transmitted very easily. The clinical signs were usually sneezing and coughing.

CONCURRENT INFECTIONS

There is some question as to whether other viruses can predispose to SIV, but experimentally infection with PRCV and H1N1 or H3N2 SIV has not shown this. Pigs with both M. hyopneumoniae and SIV coughed more and had more pneumonia than either of the two agents on their own. Preinfection with M. hyopneumoniae modifies the outcome of infection with SIV H1N1 but not H1N2. The H1N2 was more pathogenic than the H1N with an earlier shedding and greater spread in the lungs. The M. hyo and H1N1 seemed to act synergistically, but the M. Hyo and H1N2 seemed to compete because H1N2 appeared to eliminate M. hyo in the caudal lobes.157 The occurrence of SIV in pigs presents opportunities for an increased impact of bacterial infections such as H. parasuis (HPS). It has been shown that coinfection between H3N2 and both a virulent and nonvirulent strain of HPS and porcine bone marrow dendritic cells was heightened because it raised the levels of Il-1β, TNF-α, IL-6, IL-12, and IL-10 compared with SIV or mock infections.158 With the virulent strain of HPS, Il-12 and IFN-α increased differentially.

1082

Chapter 12 

■ 

Diseases of the Respiratory System

CLINICAL PATHOLOGY

VetBooks.ir

Experimental Infections Following experimental H1N1 infection, it was found that IFN-α, IL-6, IL-1, and TNF-α peaked in bronchoalveolar lavage fluid (BALF) at 24 to 30 hours postinfection, when virus titers and the severity of the clinical signs were maximal.159 Serum cytokine concentrations were not detectable or 100-fold lower than the BALF readings, but IFN-γ and IL-12 in serum followed the lavage pattern. The acute-phase protein(APP), C-reactive protein, and haptoglobin were raised 24 hours after the cytokine response, and the lipopolysaccharide binding protein only increased in the BALF. The findings suggested that IFN-α and IL-12 play an important part in the pathogenesis of SIV and that APPs are induced by cytokines.164 Acute-phase proteins and serum amyloid were raised when pigs were simultaneously infected with H1N1 virus and P. multocida.160 Experimental infections with the human 1918 pandemic influenza virus produced only a mild disease and pigs, and they did not become moribund, whereas in other mammalian species the effects were lethal.161 The findings suggested that the virus entered the swine population from humans and then established the classical H1N1 lineage in pigs. Experimental infection with H1N1 European swine influenza virus protects pigs from infection with the 2009 pandemic H1N1 human virus.162 Experimental infections with the U.S. isolates of the p(H1N1) 2009 were described,163 and all the pigs developed clinical signs similar to those induced by endemic SIV viruses. Within 24 hours of the onset of clinical signs there is a switch of cells in the bronchial lavage from macrophages to over 50% neutrophils. Serologic Tests After infection has ceased to circulate in the herd, SIV AB could still be demonstrated after 28 months postinfection. It is extremely important to make sure that the antigens that are used in the serologic tests are contemporary to the viral strains that may be found in the country. Diagnosis of acute SIV infections requires the use of paired serum samples. The hemagglutination inhibition test has been the recommended test for many years and still remains so. However, it is tedious and has only moderate sensitivity but high specificity. It has been adapted and modified. One HI test for H1N1 will detect other H1N1 strains, but this is not true for H3N2 when the Midwest strains are compared with the North Carolina strains because they differ considerably. Above 1 : 80 is usually considered positive, and within 5 to 7 days the titers may reach 1 : 320 to 1 : 640 by 2 to 3 weeks postinfection. An ELISA-based test is now available to estimate the hemagglutination titer and can be used at the herd

screening level.164 Antiinfluenza A nucleoprotein antibodies have been detected in pigs using a commercial ELISA developed for avian species.165 Detection of Virus Virus is likely to be found in the nasopharyngeal area during the acute phase of the disease. Swabs should be taken on Dacron, placed in transport medium, and stored at 4° C for no more than 48 hours; if storage will be longer, samples should be frozen at −70° C (−94 F). Viruses can also be isolated from trachea or lung tissues of pigs. They can be grown in hens’ eggs or increasingly in tissue culture. Samples need to be cool and moist. The virus is then detected by hemagglutinating activity in egg fluids about 5 days after inoculation. There are some strains that may not grow in hens’ eggs or require more than one cell line to isolate and identify the virus, which may require 1 to 2 weeks. Oral Fluids Pen-based oral fluids provide an easy, effective, and safe collection method for the detection of SI with rapid testing methods, such as RT-PCR.166 Virus isolation from nasal swabs was more successful than using oral fluids.167 The sensitivity of oral fluids for detecting influenza A virus in populations of vaccinated and nonvaccinated pigs has been described. The overall sensitivity of oral fluids was 80%, and virus was isolated from 51% of RRT-PCR positive oral fluids. The method can detect SIV even when pen prevalence is low and when pigs have been vaccinated.168 Antigen Detection A PCR test can be used to detect virus in nasal swab specimens and gives results similar to virus isolation. Recently a gelbased multiplex RT-PCR assay was developed to detect H1 and H3 subtypes of SIV. An RT-n-PCR for the identification of SIV in clinical samples has been described.71 A realtime RT-PCR assay for differentiating the pandemic H1N1 2009 pandemic from SIVs has also been described.170 A real-time RT-PCR has been developed for the detection of p(H1N1)2009 and European SIV A infections.171 A real-time RT-PCR for pandemic influenza A virus (H1N1) 2009 matrix gene has been described.172 A multiplex RT-PCR assay for differentiating European SIV subtypes H1N1, H1N2, and H3N2 has been described169,173 and used in North American pigs.174 Loop-mediated isothermal amplification has been used for the rapid and specific detection of H3 SIV.175 There are rapid detection methods for the 2009p(H1N1) using multiplex rtRTPCR.176,177,222 The virus can be detected by direct immunofluorescence of lung tissue or lavage fluids.

Immunohistochemistry on fixed tissue is also useful. The positivity is mainly in the bronchial and bronchiolar epithelial cells and less intense in the interstitial cells and alveolar macrophages.

NECROPSY FINDINGS

Swelling and marked edema of cervical and mediastinal lymph nodes are evident. There is congestion of the mucosae of the pharynx, larynx, trachea, and bronchi. A tenacious, colorless, frothy exudate is present in the air passages. Copious exudate in the bronchi is accompanied by collapse of the ventral parts of the lungs. This atelectasis is extensive and often irregularly distributed, although the apical and cardiac lobes are most affected, and the right lung more so than the left. It may reach 50% by 4 to 5 days postinfection. The affected tissue is clearly demarcated, dark red to purple, and often reminiscent of enzootic pneumonia. Surrounding the atelectatic areas the lung is often emphysematous and may show many petechial hemorrhages. Histologically, in acute swine influenza the major feature is necrotizing bronchiolitis. There is a suppurative bronchiolitis and widespread interstitial pneumonia characterized by the early appearance of neutrophils followed by the accumulation of macrophages and mononuclear cells in the alveolar walls. After a few days there is a peribronchial and peribronchiolar infiltration of lymphocytes. In the variant of H1N1 swine influenza in Canada, there is more diffuse damage to the respiratory epithelium, resulting in firm to meaty lungs that appear thymus-like on cut surface. Microscopically, there is marked proliferation of type II pneumocytes, in addition to the presence of macrophages and necrotic inflammatory cells in the alveoli. The influenza type A virus can be demonstrated by indirect immunofluorescence staining using monoclonal antibody directed to certain protein parts of the human type A influenza virus. The influenza type A virus can be detected and differentiated from the virus of porcine reproductive and respiratory syndrome in formalin-fixed, paraffin-embedded lung tissue using immunogold staining. Samples for Confirmation of Diagnosis These are best collected from animals with high fevers and clear nasal discharge. Most pigs may excrete virus for 5 to 7 days postinfection, but the peak load may be around 24 hours postinfection • Histology—formalin-fixed lung, trachea, turbinate (LM, IHC). After 72 hours there is little IFA or IHC positivity. Histopathology may help in the diagnosis for 2 weeks postinfection. • Virology—nasopharyngeal swab in viral transport media; lung and

Diseases of the Swine Respiratory Tract

VetBooks.ir

trachea (ISO, FAT, PCR) fresh chilled but not frozen. Keep cool. Do not use cotton. DIFFERENTIAL DIAGNOSIS The explosive appearance of an upper respiratory syndrome, including conjunctivitis, sneezing, and coughing, with a low mortality rate, serves to differentiate swine influenza from the other common respiratory diseases of swine. Enzootic pneumonia of pigs is most commonly confused with swine influenza, but it is more insidious in its onset and chronic in its course. Hog cholera is manifested by less respiratory involvement and a high mortality rate. Inclusion-body rhinitis in piglets may resemble swine influenza quite closely. Atrophic rhinitis has a much longer course and is accompanied by characteristic distortion of the facial bones.

TREATMENT No specific treatment is available. Treatment with penicillin, sulfadimidine, or, preferably, a broad-spectrum antibiotic may be of value in controlling possible secondary invaders. The provision of comfortable, well-bedded quarters, free from dust, is of major importance. Clean drinking water should be available, but feed should be limited during the first few days of convalescence. Medication of the feed or water supplies with a broadspectrum antibiotic for several days is a rational approach to minimizing secondary bacterial pneumonia. A novel monoclonal antibody was shown to be effective against lethal challenge with swine lineage and 2009 pandemic H1N1 influenza viruses.178

CONTROL

Treatment of human influenza is possible with oseltamivir, but some viruses have become resistant; however, there is no evidence that natural oseltamivir resistance in swine and wild waterbirds is common.179 There are only two options: vaccination and biosecurity. Biosecurity is difficult because there is always the possibility of aerosol infections and wild fowl/poultry infections It should be aimed at preventing transmission from people to pigs and vice versa. Eradication following herd closure and partial depopulation has been achieved.180 There was no introduction of replacement animals, replacement gilt deliveries were seronegative and went to quarterly instead of monthly, and the nursery was totally depopulated along with the finishing sites once shedding had finished.181 The perceptions of the pig producers in Australia in response to the occurrence of

the pandemic182 suggests that ongoing communications about biosecurity are very important when new outbreaks occur. Vaccination against swine influenza in a herd experiencing an outbreak of PCVAD is of questionable value.183 A study of vaccination in pigs infected with PRRS at the time of vaccination against SIV showed increased levels of macroscopic and microscopic lesions and also increased clinical disease and shedding of the virus.184 All-in, all-out systems may remove infection with each group of pigs, and the subsequent disinfection may wipe out the virus. Good housing and protection from inclement weather help to prevent the occurrence of severe outbreaks. Once the disease has appeared on a unit, there is little that can be done to prevent spread to other pigs. Recovered animals are immune to subsequent infection for up to 3 months. The air filtration systems proposed for PRRSV and M. hyopneumoniae185 may also be able to control SIV.

VACCINATION

Whole inactivated virus may not be the best adjuvant for the induction of cross-reactive cellular and mucosal immunity against antigenic variants. Live attenuated vaccines could prime pigs for better cross-reactivity. One method of achieving this is to use truncation of the NS1 gene200 that encodes an immune-modulating interferon antagonist. It replicates poorly but elicited neutralizing serum antibodies and mucosal antibodies and provided robust protection against homologous challenge given a single intranasal (IN) application. These vaccines provide in a single IN dose a better protection than an inactivated vaccine given intramuscularly (IM). A concern with inactivated adjuvanted vaccines is the phenomenon of vaccine-associated enhanced respiratory disease.186,187 Another obstacle is the presence of maternally derived immunity. It can reduce clinical disease, but passive antibodies are less effective in blocking viral shedding from the upper respiratory tract because the main Ig in colostrum is IgG. Pigs with maternally derived antibodies have suppressed adaptive antibody responses to homologous infection or vaccination. This interference affects IgM and HI titers in serum or nasal mucosa. The cellular response is less susceptible to maternally derived antibodies. The perception is that live attenuated IN vaccines are less likely to be interfered with by MDA.188 Virus transmission is reduced in neonatal pigs with homologous maternal immunity compared with seronegative neonatal pigs and pigs with heterologous maternal immunity.189 Vaccine development has been described.190 The genetic homology of the vaccine and the challenge virus is not the ultimate

1083

predictor for swine influenza vaccine performance.191 Vaccination with currently approved commercial vaccines in the United States did not fully prevent transmission, but certain vaccines may provide a benefit by limiting shedding, transmission, and zoonotic spillover at agricultural fairs.192 Vaccination decreases lesions and clinical signs and may eliminate virus shedding.193 Vaccines may well reduce transmission but do not eliminate infection.194 In the United States, a large number of producers vaccinate sows (~67%), and many vaccinate weaner pigs (20%). A large proportion vaccinated breeding pigs with autogenous vaccines, not commercial vaccines, and these by law are prepared by inactivating virus cultures. The main reason is that commercial vaccines are not upgraded fast enough. Vaccines (1) need to be developed quickly to keep pace with the virus changes, (2) need to have better cross-protection against new isolates, and (3) need to be able to overcome maternal antibody, which may negate vaccine use. Vaccines may use only one or two circulating strains of H3N2 in the vaccine, but the wide variation in H3N2 present in the swine population may mean that only a small percentage of currently circulating strains may be protected against by the current vaccine194 and that regular challenge studies may be necessary to determine the effectiveness of vaccines. Vaccination with influenza A virus decreased transmission rates in pigs,195 but it was not completely prevented when a heterologous vaccine was used.

INACTIVATED VACCINES

Inactivated whole-virus vaccines have limited ability or complete failure to protect against homologous challenge and even poorer cross-protection to heterologous strains.196 They can stimulate both humoral and cellular immunity.197 A trivalent inactivated swine flu vaccine was shown to be protective for all three strains (H1N1, H1N2, and H3N2).198 Inactivated vaccines from U.S. viruses and the new pandemic showed partial protection, but none was able to prevent all shedding or clinical disease.199

MODIFIED LIVE

Modified live vaccines or vectored subunit vaccines induce a balanced immune response (humoral and cell -mediated) and will improve homologous and heterologous protection. All vaccinated pigs developed a significant level of HI titer and serum IgG and IgA antibodies.200,201 A modified live vaccine as a master donor strain has been developed for the 2009 pandemic virus,202 and a pandemic virus vaccine was developed that was superior to commercial vaccines.203

1084

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

Adjuvanted and nonadjuvanted A(H1N1) pdm/09 influenza vaccines were shown to produce strong antibody responses and included high levels of specific IgG1 and HI titers to H1 virus. The adjuvanted vaccines produced a greater response.204 An eight-segment SIV with H1 and H3 was found to be attenuated and protective against both H1N1 and H3N2 subtypes in pigs.205 Vaccines, both commercial inactivated and adjuvanted SIV for IM use, are available in the United States and Europe. Active immunization occurs in the face of maternal derived antibody when titers are less than 10 for H1N1 and less than 40 for H3N2. Some of the vaccines contain the original H1N1 viruses, but others such as those used in the United States, contain a monovalent H1N1 virus. Following the outbreaks of H3N2 in the United States in 1998, both monovalent and bivalent H1N1/H3N2 SIV vaccines became available. Autogenous vaccines are used in the United States. In Europe, although the viruses have changed, the old vaccines are still used because they produce high antibody titers. There is a need to add H1N2 to the vaccines, however, because there is no cross protection between the European H1N2 and H1N1 and H3N2 viruses and because it was shown that there is no current vaccine protection against H1N2. There is evidence from the United States showing that there is cross protection with the U.S. strain of H3N2 for H1N2 infections. Most animals with titers greater than160 are probably protected against viral replication in the lungs and disease. Sow vaccination is important in controlling infection in suckling pigs and often controls the infection in nursery pigs. Intranasal or IN/IM vaccination of pigs with formalin-inactivated SIV induces very specific IgM, IgG, and IgA antibodies in their nasal secretions and sera, resulting in complete protection. A recent trial of a new H1N1/H3N2 vaccine was successful, with reduced viral shedding and reduced clinical signs and pneumonia. Experimental vaccines continue to be produced, including a human adenovirus 5 recombinant expressing the hemagglutinin and nucleoprotein of H3N2 SIV that has been used experimentally to provide protection against challenge with H3N2. Complete protection was shown by lack of nasal shedding and by lack of lung lesions following subsequent challenge. A DNA vaccine elicited robust serum antibody and cellular responses after three immunizations and conferred significant protection against influenza virus challenge.206 Vaccination with human adenovirus vector vaccines has been shown to induce both cell-mediated and humoral immunity, making them more effective than inactivated vaccines and nearly as good as live vaccines.

They can also prime the immune response in the presence of maternal antibody.207 Recently an avian-like H1N1 influenza virus was shown to be able to transmit efficiently through four pairs of vaccinated pigs at antibody levels that were thought to be protective.208 Immunity induced by infection with European avian-like H1N1 SIV affords protection for pigs against North American SIVs with a classical H1 and possibly also protects against the pH1N1.209 Pandemic (H1N1)2009 influenza viruslike particles are immunogenic.210 The vaccinated pigs were protected and showed reduced lung lesions, reduced viral shedding, and inhibition of viral replication in the lungs.

NEWER OPTIONS

Elastase-dependent SIV mutants can be used as live-virus vaccines against swine influenza in pigs.211,212 Use of the M2 conserved matrix protein may have potential as a vaccine but requires an immune response to the HA protein to reduce shedding.213 Replicon particle vaccine protects swine against influenza.214-216 Vaccination with NS1-truncated H3N2 SIV primes T cells and confers cell-mediated cross-protection against a H1N1 heterosubtypic challenge in pig.217 In addition, there was a significantly lower level of Th1associated cytokines in infected lungs. A similar vaccine can be used to differentiate between infected and vaccinated animals.218 FURTHER READING Ma W, Richt JA. Swine. Influenza vaccines: current status and future perspectives. Anim Hlth Res Rev. 2010;11:81-96. Torremorell M, et al. Transmission of influenza A virus in pigs. Transbound Emerg Dis. 2012;59(suppl 1):68-84. Vincent A, et al. Swine influenza viruses: a North American perspective. Adv Virus Res. 2008;72:127-154.

REFERENCES

1. Jung K, Song DS. Vet Rec. 2007;161:104. 2. Neumann G, Kawaoka Y. Emerg Infect Dis. 2006;12:881. 3. Morens DM, Taubenberger JK. Influenza Other Respir Viruses. 2010;4:327. 4. Parrish CR, et al. Microbiol Mol Biol Rev. 2008;457-470. 5. Taubenberger JK, Morens DM. Rev - Off Int Epizoot. 2009;28:187. 6. Song DS, et al. Virus Res. 2007;125:98. 7. Medina RA, Garcia-Sastre A. Natl Rev Microbiol. 2011;9. 8. Rossman JS, Lamb RA. Virology. 2011;411:229. 9. Taubenberger JK, Kash JC. Cell Host Microbe. 2010;7:440. 10. Vincent Al, et al. Adv Virus Res. 2008;72:127-154. 11. Conenello GM, Palese P. Cell Host Microbe. 2007;2:207. 12. Wolf YI, et al. Biol Direct. 2006;1:34. 13. Rambaut A, et al. Nature. 2008;453:615. 14. Lycett SJ, et al. J Gen Virol. 2012;93:2326.

15. Memoli MJ, et al. Virology. 2009;393:338. 16. Yassine HM, et al. Vet Microbiol. 2009;139:132. 17. Karasin AL, et al. J Clin Microbiol. 2006;44:1123. 18. Xu M, et al. Vet Microbiol. 2011;147:403. 19. Starick E, et al. Influenza Other Respir Viruses. 2011;5:276. 20. Takemae N, et al. Influenza Other Respir Viruses. 2008;2:181. 21. Lloyd LE, et al. Influenza Other Respir Viruses. 2011;5:3570. 22. Yu H, et al. Virus Res. 2009;140:85. 23. Nfon CK, et al. J Virol. 2011;85:8667. 24. Howden KJ, et al. Can Vet J. 2009;50:1153. 25. Forgie SE, et al. Clin Infect Dis. 2011;52:10. 26. Brookes SM, et al. PLoS ONE. 2010;5:39068. 27. Welsh MD, et al. Vet Rec. 2010;166:642. 28. Irvine RM, Brown IH. Vet Rec. 2009;164:577. 29. Grontvedt CA, et al. Prev Vet Med. 2013;110:429. 30. Garten RJ, et al. Science. 2009;325:197. 31. Smith GJ, et al. Nature. 2009;459:1122. 32. Lam T, et al. J Virol. 2011;85:10279. 33. Vijaykrishna D, et al. Nature. 2011;473:519. 34. Gray GC, Baker WS. Clin Infect Dis. 2011;52:19. 35. Brookes SM, et al. Vet Rec. 2011;164:760. 36. Furuse Y, et al. Virology. 2010;405:314. 37. Starick E, et al. J Gen Virol. 2012;93:1658. 38. Banyai K, et al. J Virol. 2012;86:13133. 39. Markowska-Daniel I, et al. Bull Vet Inst Pulawy. 2013;57:293. 40. Weingartl HM, et al. J Virol. 2010;84:2245. 41. Vijaykrishna D, et al. Science. 2010;328:1529. 42. Howard WA, et al. Emerg Infect Dis. 2011;17:1049. 43. Tremblay D, et al. J Clin Microbiol. 2011;49:4386. 44. Moreno A, et al. Vet Microbiol. 2011;149:472. 45. Kitikoon P, et al. Virus Genes. 2011;43:1. 46. Kitikoon P, et al. J Virol. 2012;86:6804. 47. Kitikoon P, et al. J Gen Virol. 2013;94:1236. 48. Pereda A, et al. Influenza Other Respir Viruses. 2011;5:409. 49. Starick E, et al. J Gen Virol. 2011;92:1184. 50. Zhu H, et al. J Virol. 2011;85:10432. 51. Ducatez MF, et al. Emerg Infect Dis. 2011;17:1624. 52. Hiromoto Y, et al. Virus Res. 2012;169:175. 53. Ali A, et al. Vet Microbiol. 2012;158:60. 54. Trebbien R, et al. Virol J. 2013;10:290. 55. Olsen CW, et al. Emerg Infect Dis. 2006;12:1132. 56. Gramer MR, et al. Can J Vet Res. 2007;71:201. 57. de Jong JC, et al. J Virol. 2007;81:4315. 58. Cappuccio JA, et al. J Gen Virol. 2011;92:2871. 59. Nfon C, et al. Transbound Emerg Dis. 2011;58:394. 60. Liu Q, et al. Arch Virol. 2012;157:555. 61. Nelson MI, et al. J Virol. 2012;86:8872. 62. Su S, et al. J Virol. 2012;17:9542. 63. Kim S-H, et al. Arch Virol. 2013;158:2351. 64. Nidom CA, et al. Emerg Infect Dis. 2010;16:1515. 65. Lipatov AS, et al. PLoS Pathog. 2008;4:e1000102. 66. Kwon TY, et al. Vet Microbiol. 2007;153:393. 67. Killian Ml, et al. Avian Dis. 2011;55:611. 68. Ma W, et al. Proc Natl Acad Sci United States. 2007;104:20940. 69. Shin J-Y, et al. J Clin Microbiol. 2006;44:3923. 70. Moreno A, et al. Vet Microbiol. 2009;138:361. 71. Octaviani CP, et al. J Virol. 2010;84:10918. 72. Takano R, et al. Arch Virol. 2009;154:677. 73. Lee JH, et al. J Virol. 2009;83:4205. 74. Song X-H, et al. Zoonoses Public Health. 2010;57:291. 75. Londt BZ, et al. Vet Microbiol. 2013;162:944. 76. Zhao G, et al. Res Vet Sci. 2013;95:434. 77. Kwon TY, et al. Vet Microbiol. 2011;1253:393. 78. Cong YL, et al. J Gen Virol. 2007;88:2035. 79. Sun Y, et al. Vet Microbiol. 2010;146:215. 80. Jones JC, et al. J Virol. 2013;87:12496. 81. Zhu H, et al. Science. 2013;341:183. 82. Wang N, et al. J Virol. 2012;86:13866.

Diseases of the Swine Respiratory Tract

VetBooks.ir

83. Hofshagen M, et al. Euro Surveill. 2009;14:19406. 84. Liu W, et al. Vet J. 2011;187:200. 85. Mastin A, et al. PLoS Curr. 2011;3:RRN1209. 86. Van Reeth K, et al. Influenza Other Respir Viruses. 2008;2:99. 87. Markowska-Daniel I, Kowalcczyk A. Med Wet. 2007;61:669. 88. Jung K, et al. Prev Vet Med. 2007;79:294. 89. Pascua PN, et al. Virus Res. 2008;138:43. 90. Suriya R, et al. Zoonoses Public Health. 2008;55:342. 91. Poljak Z, et al. Can J Vet Res. 2008;72:7. 92. Poljak Z, et al. Prev Vet Med. 2008;83:24. 93. Mancini D, et al. Virus Rev Res. 2006;11:39. 94. De Vleesschauwer A, Van Reeth K. Vet Microbiol. 2010;146:340. 95. Kyriakis S, et al. Zoonoses Public Hlth. 2011;58: 93. 96. Markowska-Daniel I, Kowalczyck A. Med Wet. 2007;61:669. 97. Howden KJ, et al. Canad Vet J. 2009;50:1153. 98. Hofshagen M, et al. Euro Surveill. 2009;14:19406. 99. Moreno A, et al. Open Virol J. 2010;4:52. 100. Pasma T, Joseph T. Emerg Infect Dis. 2010;16:706. 101. Pereda A, et al. Emerg Infect Dis. 2010;16:304. 102. Song MS, et al. J Clin Microbiol. 2010;48:3204. 103. Sreta D, et al. Emerg Infect Dis. 2010;16:1587. 104. Forgie SE, et al. Clin Infect Dis. 2011;52:10. 105. Loeffen WLA, et al. Vet Microbiol. 2009;137:45. 106. Mastin A, et al. PLoS Curr. 2011;3:RRN1209. 107. Simon-Grife M, et al. Vet Microbiol. 2011;149:56. 108. Larsen LE, et al. Proc 21st Int Pig vet Soc Cong. 2010;80. 109. Ma W, et al. J Gen Virol. 2012;93:1261. 110. Lowen AC, et al. PLoS Pathog. 2007;3:1470. 111. Zhang H, et al. Virol J. 2013;10:204. 112. Brown JD, et al. Avian Dis. 2007;51:285. 113. Brown JD, et al. Vet Microbiol. 2009;136:20. 114. Tiwari A, et al. Avian Dis. 2006;50:284. 115. Thomas Y, et al. Appl Environ Microbiol. 2008;74:3002. 116. Brookes SM, et al. Vet Rec. 2009;164:760. 117. Lange E, et al. J Gen Virol. 2009;90:2119. 118. Tellier R. Emerg Infect Dis. 2006;12:1657. 119. Lowen AC, et al. Proc Natl Acad Sci United States. 2006;103:9988. 120. Mubareka SJ, et al. Infect Dis J. 2009;199:858. 121. Yee KS, et al. Avian Pathol. 2009;38:59. 122. Yee KS, et al. Virology. 2009;394:19. 123. Romijn PC, et al. Vet Rec. 2009;124:224. 124. Sawabe K, et al. Am J Trop Med Hyg. 2006;75:327. 125. Sawabe K, et al. J Med Entomol. 2009;46:852. 126. Nelson MI, et al. PLoS Pathog. 2011;7:e1002077. 127. Vijaykrishna D, et al. Nature. 2011;473:519. 128. Kyriakis CS, et al. Emerg Infect Dis. 2010;16:96. 129. Kitikoon P, et al. Vet Immunol. 2006;112:117. 130. Van Reeth K, et al. Vaccine. 2009;27:6330. 131. Myers KP, et al. Clin Infect Dis. 2007;44:1084. 132. Gerloff NA, et al. Emerg Infect Dis. 2011;17:403. 133. Myers KP, et al. Clin Infect Dis. 2006;42:14. 134. Terebuh P, et al. Influenza Other Respir Viruses. 2010;4:387. 135. Gray GC, et al. Vaccine. 2007;25:4376. 136. Beaudoin A, et al. Influenza Other Respir Viruses. 2010;4:163. 137. Shinde V, et al. N Engl J Med. 2009;360:2616. 138. Bowman AS, et al. Emerg Infect Dis. 2012;18: 1945. 139. Vincent AL, et al. Vet Microbiol. 2009;137:51. 140. Nelli RK, et al. Vet Res. 2010;6:4. 141. Poucke SGM, et al. Virol J. 2010;7:38. 142. Trebbien R, et al. Virol J. 2011;8:434. 143. Detmer SE, et al. Vet Pathol. 2013;50:648. 144. Charley B, et al. Ann New York Acad Sci. 2006;1081:130.

145. Michael B, et al. Viruses. 2011;3:312. 146. Mussa T, et al. Virology. 2011;420:125. 147. Mussa T, et al. Vet Immunol Immunopathol. 2013;154:25. 148. Barbe F, et al. Res Vet Sci. 2010;88:172. 149. Safronetz D, et al. J Virol. 2011;85:1214. 150. Krumbholz A, et al. Med Micrbiol Immunol. 2011;200:69. 151. Ma W, et al. J Virol. 2011;85:e11626. 152. Munster VJ, et al. Science. 2009;325:481. 153. Maines TR, et al. Science. 2009;325:484. 154. Huang Y, et al. Arch Virol. 2013;158:2267. 155. Simon-Grife M, et al. Vet Res. 2012;43:24. 156. Williamson SM, et al. Vet Rec. 2012;171:271. 157. Deblanc C, et al. Vet Microbiol. 2012;157:96. 158. Mussa T, et al. Vet Res. 2012;43:80. 159. Barbe F, et al. Vet J. 2013;187:48. 160. Pomorska-Mol M, et al. Vet Res. 2013;9:14. 161. Weingartl HM, et al. J Virol. 2009;83:4287. 162. Busquets N, et al. Vet Res. 2010;41:74. 163. Vincent AL, et al. Influenza Other Respir Viruses. 2010;4:53. 164. Barbe F, et al. J Vet Diag Invest. 2009;21:88. 165. Ciacci-Zanella JR, et al. J Vet Diag Invest. 2010;22:3. 166. Detmer SE, et al. J Vet Diag Invest. 2011;23:241. 167. Goodell CK, et al. Vet Microbiol. 2013;166:450. 168. Romagosa A, et al. Influenza Other Respir Viruses. 2012;6:110. 169. Kowalczyk A, et al. Med Wet. 2007;63:810. 170. Hiromoto Y, et al. J Virol Meth. 2010;170:169. 171. Slomka MJ, et al. Influenza Other Respir Viruses. 2010;4:277. 172. Lorusso A, et al. J Virol Meth. 2010;164:83. 173. Chiapponi C, et al. J Virol Meth. 2012;184:117. 174. Nagarajan MM, et al. J Vet Diag Invest. 2010;22:402. 175. Gu H, et al. J Appl Microbiol. 2010;108:1145. 176. Harmon K, et al. Influenza Other Respir Viruses. 2010;4:405. 177. Hofmann B, et al. Berl Munch Tierartzl Wschr. 2010;123:286. 178. Shao H, et al. Virology. 2011;417:379. 179. Stoner TD, et al. J Virol. 2010;84:9800. 180. Torremorell M, et al. Vet Rec. 2009;165:74. 181. Schafer N, Morrison RBJ. Sw Hlth Prod. 2007;15:152. 182. Hernandez-Jover M, et al. Prev Vet Med. 2012;106:284. 183. Poljak Z, et al. Can J Vet Res. 2010;74:108. 184. Kitikoon P, et al. Vet Microbiol. 2009;139:235. 185. Dee S, et al. Virus Res. 2010;154:177. 186. Gauger PC, et al. Vaccine. 2011;29:2712. 187. Vincent AL, et al. Vet Microbiol. 2008;126:310. 188. Vincent AL, et al. J Virol. 2012;86:10597. 189. Allerson M, et al. Vaccine. 2013;31:500. 190. Chen Q, et al. Anim Hlth Res Rev. 2012;13:181. 191. Kyriakis CS, et al. Vet Microbiol. 2010;144:67. 192. Loving CL, et al. J Virol. 2013;87:9895. 193. Lee JH, et al. Can J Vet Res. 2007;71:207. 194. Gramer MR, et al. Can J Vet Res. 2007;71:201. 195. Romagosa A, et al. Vet Res. 2011;42:120. 196. Vincent AL, et al. Vet Microbiol. 2008;126:310. 197. Platt R, et al. Vet Immunol Immunopathol. 2011;142:252. 198. Durrwald R, et al. Tieratl Prax. 2009;37:103. 199. Vincent AL, et al. Vaccine. 2010;28:2782. 200. Richt JA, et al. J Virol. 2006;80:11009. 201. Vincent AL, et al. Vaccine. 2007;25:2999. 202. Pena L, et al. J Virol. 2011;85:456. 203. Loeffen WLA, et al. Vet Microbiol. 2011;152:304. 204. Lefevre EA, et al. PLoS ONE. 2012;7:e32400. 205. Masic A, et al. J Virol. 2013;87:10114. 206. Gorres JP, et al. Clin Vaccine Immunol. 2011;18:1987.

1085

207. Wesley RD, Lager KM. Vet Microbiol. 2006;118:67. 208. Lloyd LE, et al. Influenza Other Respir Viruses. 2011;5:357. 209. De Vleeschauwer AR, et al. Influenza Other Respir Viruses. 2010;5:115. 210. Pyo H-M, et al. Vaccine. 2012;30:1297. 211. Masic A, et al. J Virol. 2009;83:10198. 212. Masic A, et al. Vaccine. 2010;28:7098. 213. Kitikoon P, et al. Vaccine. 2010;28:523. 214. Erdman MM, et al. Vaccine. 2010;28:594. 215. Bosworth B, et al. Comp Immunol Microbiol Infect Dis. 2010;33:e99. 216. Vander Veen RL, et al. Vet Rec. 2013;doi:1136/ vr.101741. 217. Kappes MA, et al. Vaccine. 2012;30:280. 218. Richt JA, et al. J Virology. 2006;80:11009. 219. Kobayashi M, et al. Emerg Infect Dis. 2013;19:1972. 220. He LO, et al. Arch Virol. 2013;158:2531. 221. Londt B, et al. Virus Res. 2013;178:383. 222. Ma W, et al. Influenza Other Respir Viruses. 2010;4:397.

PORCINE RESPIRATORY CORONAVIRUS Infection with coronavirus causes a rapid seroconversion to some of the tests for TGE and is responsible for “vaccinating” large populations of pigs worldwide against the threat of TGE. This has coincided with the great reduction in TGE in most countries. It was first identified in Belgium in 1986 and since then has spread worldwide.

ETIOLGY

The virus is very similar to TGE, and the major difference is a 621- to 628-base-pair deletion in the S protein gene causing a truncated S protein and loss of the ability of the TGE to bind sialic acid. It has a tropism for the respiratory tract. It is one of the four swine coronaviruses and is a mutant of TGE, first isolated in 1984. The virus has been fully or partially sequenced and has 96% to 98% homogeneity with TGE.1 Lipoteichoic acid from S. aureus exacerbates respiratory disease in PRCV-infected pigs,2 and coinfection with B. bronchiseptica is reported.3 PRRSV-induced immunosuppression exacerbates the inflammatory response to PRCV in pigs.4 PRCV-infected pigs produce antibodies that neutralize TGE virus.

EPIDEMIOLOGY

The virus distribution is affected by the season and the density of pig farms, and in a dense area there is rapid local spread probably by aerosol. The virus infects pigs of all ages by contact or airborne transmission and in areas of high density can probably spread several kilometers. The virus circulates in the herd, infects pigs less than 10 to 15 weeks of age after the maternal antibodies have declined, and becomes endemic. Experimentally, infected pigs shed virus from the nose for less than 2 weeks. The infection can be maintained in herds, cycle regularly, or appear in waves. In Europe, these waves often coincide with the rainy season. There is no evidence of fecal/oral transmission.

1086

Chapter 12 

■ 

Diseases of the Respiratory System

PATHOGENESIS

VetBooks.ir

The virus has a tremendous ability to replicate in the respiratory tract in most of the airway but rarely the alveolar macrophage.5-8 The main targets are type 1 and type 2 alveolar epithelial cells, and it induces necrosis in these cells, causing a rise in cytokines that induces a rise in nitric oxide and IFN-α. The shedding from the nose lasts 4 to 6 days. The pneumonia produced and the viral replication peak at 7 to 10 days postinoculation and then resolve with the increasing levels of neutralizing antibody.

CLINICAL SIGNS

Most infections are inapparent, but in a susceptible population there may be respiratory signs such as labored breathing and coughing, followed by depression, anorexia, and decreased growth rates.

LESIONS

The lesions are usually self-limiting. The major lesions are broncho-interstitial pneumonia with cuffing and syncytial formation from type 2 hyperplasia, followed by necrosis and lymphoid hyperplasia. Necrotic cells and inflammatory cells may obstruct the lumen of the alveoli.

DIAGNOSIS

Virus isolation in PK and swine testicle cells is necessary using nasal fluid or lung homogenates, and frequently PRCV produces syncytia in culture. Respiratory samples are required for diagnosis of PRCV. Currently, RT-PCR or qRT-PCR is needed to differentiate TGEV and PRCV. The primers target the S protein. Multiplex PCR has now been developed for TGEV, PRCV, and PEDV9 and up to eight viruses. Multiplex microarray has also been developed for the rapid differentiation of eight coronaviruses.10 Blocking ELISAs have been developed to differentiate antibodies of PRCV from TGE and should be used on a herd basis. Recently, new ELISAs have been developed that will also differentiate TGE, PRCV, and the new TGE-like coronaviruses.11,12

TREATMENT

There is no treatment for PRCV infections except supportive therapy and control of secondary infections.13

CONTROL

Neonatal pigs require 6 to 8 days after PRCV exposure to produce partial immunity to TGE. Sows naturally exposed to PRCV reinfected with PRCV during pregnancy secreted TGEV antibodies in milk and provided a high degree of protection. REFERENCES

1. Zhang X, et al. Virology. 2007;358:424. 2. Atanasova K, et al. Vet J. 2011;188:210. 3. Brockmeier SL, et al. Vet Microbiol. 2008;128:36.

4. Renukaradhya GJ, et al. Viral Immunol. 2010;23:457. 5. Atanasova K, et al. Open Vet Sci J. 2008;2:117. 6. Jung K, et al. J Virol. 2007;81:13681. 7. Jung K, et al. J Gen Virol. 2009;90:2713. 8. Jung K, et al. Vet Immunol Immunopathol. 2010;136:335. 9. Ogawa H, et al. J Virol Meths. 2009;160:210. 10. Chen q, et al. Intervirol. 2010;53:95. 11. Elia G, et al. J Virol Methods. 2010;163:309. 12. Lopez I, et al. J Vet Diag Invest. 2009;21:598. 13. Zhang X, et al. J Virol. 2008;82:4420.

infestations are asymptomatic and induce immunity against reinfection.

PATHOGENESIS

The pathogenesis is similar to that of D. viviparus. These worms may provide a route of transmission for swine influenza virus, and possibly hog cholera virus, from pig to pig, but this is unproven.

CLINICAL FINDINGS

The lungworms that infest pigs are Metastrongylus apri (M. elongatus), M. salmi, and M. pudendotectus. M. apri is the most common species, but mixed infestations are not uncommon.

Lungworm infection in pigs can cause a marked check in growth rate. The bronchitis is accompanied by sporadic bouts of a barking cough, which is easily stimulated by exercise. Pneumonia is a feature of severe cases. Fatal bronchopneumonia can occur in coinfections of porcine circovirus type 2 and Metastrongylus spp.1

LIFE CYCLE

CLINICAL PATHOLOGY

LUNGWORM IN PIGS ETIOLOGY

Adult Metastrongylus spp. appear much like D. viviparus in the bronchi of their host. Their life cycles are also similar, except that Metastrongylus spp. eggs are passed in the feces and earthworms act as intermediate hosts. Here development to infective larvae takes about 2 weeks, and transmission occurs when the earthworm is eaten by a pig.

Laboratory diagnosis is by demonstration of the characteristic eggs in feces.

NECROPSY FINDINGS

Etiology The nematode parasites Metastrongylus apri (M. elongatus), M. salmi, and M. pudendotectus.

Early lesions comprise small areas of consolidation as a result of verminous pneumonia. More chronic cases have bronchitis, emphysema, peribronchial lymphoid hyperplasia, and bronchiolar muscular hypertrophy, often accompanied by areas of overinflation. The lesions are small and discrete, appearing as grayish nodules up to 1 cm in diameter, and are present particularly at the ventral border of the diaphragmatic lobes.

Epidemiology Transmission is by ingestion of the earthworm intermediate host.

DIAGNOSTIC CONFIRMATION

SYNOPSIS

Signs Check in growth rate; barking cough. Clinical pathology Characteristic eggs in feces. Lesions Grayish nodules near the ventral border of the diaphragmatic lobes of the lung. Diagnostic confirmation Characteristic eggs in feces. Treatment Doramectin, ivermectin, fenbendazole, flubendazole, levamisole. Control Difficult, unless pigs reared on concrete.

EPIDEMIOLOGY The disease is most prevalent in pigs 4 to 6 months of age in husbandry systems that allow access to earthworms. The eggs first appear in the feces 3 to 4 weeks after infestation and at their peak reach levels of 25 to 50 eggs per gram of feces. The eggs are very resistant to cold temperatures and can survive for over 1 year in the soil. Larvae may survive in the earthworm for up to 7 years. The primary host must ingest an intermediate host to become infested, and this is an important factor influencing the spread of the disease. Once ingested the infective larvae migrate to the lungs in much the same manner as do D. viviparus larvae. Many

The Metastrongylus egg is embryonated (larvated) and has a thick shell and a wavy outline. They may be missed on routine screening as they are usually passed in small numbers and do not float well in saturated salt (NaCl) solution. A flotation fluid with a higher specific gravity should be used. There will always be a history of access to yards or paddocks where earthworms exist. DIFFERENTIAL DIAGNOSIS • Other swine pneumonias • Migrating larvae in heavy Ascaris infestation

TREATMENT TREATMENT Abamectin (0.1 mg/kg, PO) (R1) Ivermectin (0.3 mg/kg, SC) (R2) Fenbendazole (9 mg/kg, PO qd for 3 days) (R2) Flubendazole (4.0 mg/kg, PO) (R2) Levamisole (8 mg/kg, PO)

A number of anthelmintics are effective at normal pig dose rates, including abamectin,

Respiratory System Toxicoses

VetBooks.ir

ivermectin,2 doramectin, fenbendazole, and flubendazole. Levamisole (8 mg/kg) has been used in the water or feed.

CONTROL

Rearing pigs on concrete reduces the risk considerably but, in view of the longevity of the eggs and larvae in the earthworm, little can be done if pigs are kept on contaminated land. Pastures that are known to be contaminated should be left for at least 6 months before restocking, although infested earthworms may persist in hog lots for up to 4 years. FURTHER READING Roepsdorff A, Mejer H, Nejsum P, Thamsborg SM. Helminth parasites in pigs: new challenges in pig production and current research highlights. Vet Parasitol. 2011;180:72.

REFERENCES

1. Lopes WD, et al. Res Vet Sci. 2014;97:546. 2. Marruchella G, et al. Res Vet Sci. 2012;93:310.

Respiratory System Toxicoses FURAN (IPOMEANOL AND 3-METHYLINDOLE) TOXICOSIS 4-Ipomeanol (4-IPO) is a furanoterpinoid mycotoxin produced by Fusarium solani (synonym F. javanicum) and F. semitectum growing on garden refuse. It has the effect of causing lesions indistinguishable from those of atypical interstitial pneumonia. Other known causes of these lesions are 3-methylindole and the ketone produced by Perilla frutescens, Zieria arborescens, and one of the fungi Fusarium solani or Oxysporum spp. on Ipomoea batatas (sweet potatoes) tubers and tryptophan-containing plants.1 Catabolism by the fungus of phytoalexins induced in the tubers produces four closely related ipomeanols: ipomeanine (IPO), 4-ipomeanol (4-IPO), 1-ipomeanol (1-IPO), and 1,4-ipomeadiol (DIOL).2 These are not toxic until activated by pulmonary microsomal enzymes; 4-IPO and IPO are ultimately the most toxic. Experimental administration of infected potatoes to calves is associated with bronchiolitis and interstitial pneumonia. Unweaned, nursing calves may not be affected.3 Animals are exposed to these toxins in a number of ways. Cows gain access to moldy sweet potatoes by grazing plowed potato fields or being fed spoiled sweet potatoes. The toxic dose is 7.5 mg IPO/kg BW, which converts to about 6 kg of spoiled sweet potatoes per adult cow.1 The mortality rate is often high.4 Perilla mint (purple mint or beefsteak plant) is widespread in the southeastern United States4 and found in Asia and several other parts of the world.1,5 All large animal species are susceptible, but poisoning is most widely reported in cattle.

Cows are exposed by eating the leaves and seeds; toxicity is highest in the seed portion of the plant.4 In a similar fashion, tryptophan toxicosis occurs in cows grazing on lush pastures with elevated concentrations of tryptophan. Outbreaks often develop several days to a week after cows are moved from poor pastures or forage to early summer pastures with high tryptophan content in the grasses. Rumen microflora convert tryptophan to 3-methylindole, which is then activated by cytochrome p450 in the lung to a reactive compound.4 The clinical signs present in ipomeanol and 3-methylindole toxicosis are similar to acute respiratory distress syndrome and atypical interstitial pneumonia. The reactive compounds produced in the lung damage the pulmonary endothelial cells and result in acute pulmonary emphysema and edema.1,4 Affected animals have labored breathing, frequently standing with an open mouth and extended neck. Frothy foam from the nostrils or a foam-covered tongue may be present. Treatment is aimed toward reducing edema, supporting respiration, and reducing physical stress. Animals living longer than 48 hours have a good prognosis for survival.4 FURTHER READING Kerr LA, Johnson BJ, Burrows GE. Intoxication of cattle by Perilla frutescens (purple mint). Vet Hum Toxicol. 1986;28:412-416. Yokoyama MT, Carlson JR, Dickinson EO. Ruminal and plasma concentrations of 3-methylindole associated with tryptophan-induced pulmonary edema and emphysema in cattle. Am J V. 1975;36: 1349-1352.

REFERENCES

1. Parkinson OT, et al. J Vet Pharmacol Therp. 2012;35:402. 2. Chen LJ, et al. Chem Res Toxicol. 2006;19:1320. 3. Mawhinney I, et al. Cattle Pract. 2009;17:96. 4. Nicholson SS. Vet Clin North Am Food A. 2011;27:456. 5. Lee Y-J, et al. J Taiwan Agric Res. 2009;58:2114.

GALEGINE TOXICOSES Galegine, an isoprenoid guanidine, is found in the following plants: Galega officinalis: French honeysuckle1 Schoenus asperocarpus: poison sedge (Australia) S. rigens (Australia) Verbesina encelioides: crown beard (North America and Australia)1 Ingestion of galegine-containing plants is associated with a syndrome of severe dyspnea, frothing from the nose, convulsions, and sudden death in ruminants as a result of pulmonary edema with large fluid accumulations in the thoracic cavity, the result of a direct effect on pulmonary vascular permeability.1 Sheep may find access via

1087

these plants being mixed in with hay or among a standing crop. REFERENCE

1. Jai SC, et al. Indian J Trad Know. 2008;7:511.

MANURE GAS POISONING AND CONFINEMENT EFFECTS ETIOLOGY

Confinement housing of cattle and swine is accompanied by manure storage for varying periods of time, often in large holding pits under slatted floors. Oxygen is excluded from the storage so that anaerobic bacteria degrade the organic and inorganic constituents of manure, yielding hydrogen sulfide, ammonia, methane, and carbon dioxide as major gases.1,2 When diluted with water to facilitate handling, liquid manure in storage separates by gravity. The solid wastes form sediment, the lightweight particles float to the top, leaving a middle layer that is relatively fluid. Thorough remixing is necessary before pits are emptied to prevent the fluid fraction from flowing out and the solids remaining. The remixing or agitation results in the release of large quantities of toxic gases from the slurry.2 Besides the well-established gaseous toxicants listed, certain other agents with detrimental inhalation risks are present in confinement operations and have been best characterized for swine confinement operations. Total dust is a major contaminant in swine barns3 and may range from 2 to 7 mg/m. Particulates may adsorb gases and be part of the objectionable odors released and reaching neighbors near confinement operations. Respirable dusts may be 10% or more of the total dusts generated in swine barns. Such dust is contaminated with bacteria, fungi, endotoxins, and glucans.3 Dusts are primarily composed of feed or fecal material. Both endotoxins and glucans have been suggested as potential contributors to swine respiratory disease and respiratory complications for workers in swine buildings. So far, however, high mortality and acute death losses in confinement operations are most commonly caused by excessive concentrations of hydrogen sulfide and carbon dioxide, whereas subacute or chronic irritation and disease of the upper respiratory tract may also be contributed by elevated ammonia levels. Methane is explosive and may act as an asphyxiant, but is not implicated as a toxicant. Additional factors that must be considered in a differential diagnosis include possible power loss during electrical storms or equipment failure; this results in the cessation of the artificial ventilation required to cool the building and exhaust carbon dioxide from the animals’ respiration. In these situations, CO2 levels build rapidly, and environmental temperatures increase dramatically as well, especially when weather conditions

1088

Chapter 12 

■ 

Diseases of the Respiratory System

VetBooks.ir

are hot and humid.1 Acute loses from hyperthermia or heat stroke may be mistaken for manure gas poisoning.1 This is important for veterinarians because they may be called to establish a diagnosis that affects insurance claims for many thousands of dollars. Besides overheating and CO2 accumulation, electrocution should be considered whenever there are large numbers of acute losses in a confinement building.

PATHOGENESIS

The exposure of humans, cattle, and swine to high concentrations (above 700 ppm of H2S) of manure gases, particularly hydrogen sulfide, can be associated with peracute deaths in cattle and swine. Hydrogen sulfide is both an irritant and an acute toxicant. Fatal or severe exposure often is associated with respiratory distress and pulmonary edema. Exposure to low concentrations of hydrogen sulfide over long periods is thought to be associated with reduced performance in cattle and swine. At high concentrations, from 500 to 1000 ppm, carotid-body receptors are stimulated, causing rapid breathing. As high concentrations continue or increase, the respiratory center is depressed, and animals become depressed and die. High concentrations of H2S depress olfactory sensors, and the offensive rotten-egg odor is no longer detected as a warning sign. Ammonia is either an irritant or corrosive agent depending on the concentration. Ammonia combines with tissue moisture to produce ammonium hydroxide, a strong alkali capable of causing tissue necrosis.

CLINICAL FINDINGS

In acute hydrogen sulfide poisoning the animals die suddenly. Affected animals may be found dead throughout a building in various postures of lateral or sternal recumbency. There may be little or no evidence of struggle or excitement because high concentrations can be associated with nearly immediate respiratory paralysis. In acute ammonia poisoning the syndrome includes conjunctivitis, sneezing, and coughing for a few days, but pigs will soon acclimatize, after which no effects may be detectable. An increased incidence of pneumonia and reduced daily weight gains in pigs are asso­ ciated with exposure to a combination of gaseous ammonia at levels of 50 to 100 ppm and the presence of atmospheric dust in barns. Higher concentrations of ammonia (100-200 ppm) are associated with irritation to the conjunctiva and respiratory mucosa. At very high ammonia concentrations (>500 ppm), there is pharyngeal and laryngeal irritation, laryngospasm, and coughing. Concentrations above 2000 ppm can be associated with death within 30 minutes. Carbon dioxide overexposure first is associated with mild to moderate excitement, followed by depression, weakness, coma, and

death. Concentrations above 30% in air are serious, and 40% CO2 for more than a few minutes can cause death.

NECROPSY FINDINGS

In cattle that have died from acute hydrogen sulfide poisoning, lesions include pulmonary edema, extensive hemorrhage in muscles and viscera, and bilaterally symmetric cerebral edema and necrosis. Ammonia exposure results in lacrimation, conjunctivitis, corneal opacity, tracheal hyperemia or hemorrhages, and pulmonary edema. Secondary bacterial pneumonia may be evident in exposed animals. For carbon dioxide, the principal lesions are of cyanosis.

CONTROL

Production of hydrogen sulfide in manure can be inhibited by aeration using air as the oxidizing agent or the use of chemical oxidizing agents. The use of ferrous salts virtually eliminates hydrogen sulfide evolution. Adequate ventilation with all doors and windows wide open during remixing and agitation of the slurry will reduce the concentration of hydrogen sulfide to nontoxic levels. Animals and personnel should not enter closed barns when the pits are being emptied. In confinement buildings, ammonia usually does not accumulate to fatal levels, but much of the economic loss is from reduced feed consumption and possibly increased susceptibility to acute or chronic respiratory disease. Limiting protein supplementation to actual needs has been considered a means for reducing nitrogen losses and the resultant production of ammonia in feces and urine. FURTHER READING Hartung J, Phillips VR. Control of gaseous emissions from livestock buildings and manure stores. J Agr Eng Res. 1994;57:173-189. Hooser SB, et al. Acute pit gas (hydrogen sulfide) poisoning in confinement cattle. J Vet Diagn Invest. 2000;12:272-275. Radostits O, et al. Manure gas poisoning. In: Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1848.

REFERENCES

1. Barrasa M, et al. Ann Agr Environ Med. 2012;19:17-24. 2. Ni JQ, et al. Sci Total Environ. 2010;408:5917. 3. Basinas I, et al. J Expo Sci Environ Epidemiol. 2013;doi:10.1038/jes.2013.83.

PLANTS CAUSING PULMONARY DISEASE (UNIDENTIFIED TOXINS) The following plants have been associated with pulmonary disease. The toxins currently are unidentified. Dyspnea and pulmonary edema: • Glechoma hederacea (= Nepeta hederacea: ground ivy) • Gyrostemon spp.: camel poison

Pulmonary consolidation and fibrosis, characterized by dyspnea and cough (horses): • Eupatorium (= Ageratina) adenophorum: crofton weed • E. riparium: mist flower • Lactuca scariola: prickly lettuce

Neoplastic Diseases of the Respiratory Tract Neoplasms arising as a result of viral infection (nasal adenocarcinoma of sheep, ovine pulmonary adenocarcinoma) and nonneoplastic tumors (equine ethmoidal hematoma) are dealt with under those headings in this chapter.

PULMONARY AND PLEURAL NEOPLASMS Primary neoplasms of the lungs, including carcinomas and adenocarcinomas, are rare in animals and metastatic tumors also are relatively uncommon in large animals. Primary tumors reported in lungs or pleura of the farm animal species include the following: Horses • Granular cell tumors are the most common tumor arising in the pulmonary tissue of horses. • Malignant melanomas in adult gray horses • Pulmonary adenocarcinoma (either primary or as metastatic disease) • Pulmonary leiomyosarcoma • Bronchogenic carcinoma, pulmonary carcinoma, bronchogenic squamouscell carcinoma, pulmonary chondrosarcoma, and bronchial myxoma are all rare tumors in lungs of horses. • Mesothelioma arise from the visceral or parietal pleura. Cattle • Pulmonary adenocarcinoma is the most commonly reported primary lung tumor in cattle. The ultrastructure and origin of some of these have been characterized. • Lymphomatosis in young cattle may be accompanied by pulmonary localization Sheep • Ovine pulmonary adenocarcinoma (jaagsiekte sheep retrovirus) is locally common in some areas. Goats • An asymptomatic, squamous-celltype tumor, thought to be a benign papilloma, has been observed in 10 of a series of 1600 adult Angora goats. The lesions were mostly in the diaphragmatic lobes, were

Neoplastic Diseases of the Respiratory Tract

VetBooks.ir

multiple in 50% of the cases, and showed no evidence of malignancy, although some had necrotic centers. • Bronchoalveolar carcinoma not related to ovine adenocarcinoma virus is reported.1 A wide variety of tumors metastasize to the lungs, and these tumors can originate in almost any tissue or organ. A series of thoracic neoplasms in 38 horses included lymphosarcoma, metastatic renal cell carcinoma, primary lung carcinomas, secondary cell carcinoma from the stomach, pleural mesothelioma, and malignant melanoma. The etiology of the tumors is unknown in most cases, apart from those arising from viral infections. Equine granular cell tumors arise from the Schwann cells of the peripheral nervous system in the lungs. Characteristically, primary pulmonary or pleural tumors arise in middle-aged to old animals. The prevalence of these tumors is not well documented, although they are rare in abattoir studies of horses. The tumors occur sporadically, with the exception of those associated with infectious agents (bovine lymphomatosis, ovine pulmonary adenocarcinoma). The pathogenesis of pulmonary tumors includes impairment of gas exchange, either by displacement of normal lung with tumor tissue and surrounding atelectasis and necrosis or by obstruction of the large airways (e.g., granular cell tumor in horses).

CLINICAL FINDINGS

Clinical findings are those usually associated with the decrease in vital capacity of the lungs and include dyspnea that develops gradually, cough, and evidence of local consolidation on percussion and auscultation. There is no fever or toxemia, and a neoplasm can be mistaken for a chronic, encapsulated pulmonary abscess. Major clinical findings included weight loss, inappetence, and dyspnea and coughing. An anaplastic smallcell carcinoma of the lung of a 6-month-old calf located in the anterior thorax caused chronic bloat, anorexia, and loss of body weight. Some tumors, notably mesothelioma and adenocarcinoma, cause accumulation of pleural fluid. Hypertrophic pulmonary osteopathy occurs in some animals with pulmonary tumors. Ovine pulmonary adenocarcinoma can metastasize to liver, kidneys, skeletal muscle, gastrointestinal tract, spleen, skin, and adrenal glands.2 Granular cell tumors in horses present as chronic coughing and exercise intolerance in horses without signs of infectious disease. As the disease progresses, there is increased respiratory rate and effort and weight loss, suggestive of severe heaves. However, horses are unresponsive to treatment for heaves. The disease can progress to cor pulmonale and right-sided heart failure. A bronchial

mass is evident on endoscopic or radiographic examination (Figs. 12-34 and 12-35). There are no characteristic hematologic or serum biochemical changes. Hemangiosarcomas of the thoracic cavities of horses occur and are evident as excess pleural fluid with a high red blood cell count.3 Thymoma, or lymphosarcoma as a part of the disease bovine viral leukosis, is not uncommon in cattle and can resemble pulmonary neoplasm, but there is usually displacement and compression of the heart, resulting in displacement of the apex beat and congestive heart failure. The presence of

jugular engorgement, ventral edema, tachycardia, chronic tympany, and hydropericardium can cause a mistaken diagnosis of traumatic pericarditis. Mediastinal tumor or abscess (cranial thoracic masses) can have a similar effect. Metastasis to the bronchial lymph nodes can cause obstruction of the esophagus with dysphagia, and in cattle chronic ruminal tympany. This tumor is also common in goats, many of which show no clinical illness. Radiographic or ultrasonographic examination is useful in demonstrating the presence of a mass in the lungs or thorax. Endoscopic examination is useful for detection of tumors that invade the larger airways, such as granular cell tumors of horses. Thoracoscopy and pleural biopsy can be useful in the diagnosis of lesions at the pleural surfaces. The nature of the tumor can sometimes be determined by examination of pleural fluid, into which some tumors shed cells, or of tumor tissue obtained by biopsy. Examination of pleural fluid for the presence of tumor cells is not very sensitive because many tumors do not shed sufficient numbers of cells to be detectable, but it is quite specific in that detection of abnormal cells is diagnostic.

TREATMENT

Fig. 12-34  Endoscopic view of a granular cell tumor in a horse.

1089

There is no effective treatment, with the exception of resection of localized tumors. Granular cell tumors in horses have been successfully treated by lung resection or transendoscopic electrosurgery.4,5

Fig. 12-35  Lateral thoracic radiograph of an adult horse demonstrating presence of a granular cell tumor (outline by black arrows).

1090

Chapter 12 

■ 

Diseases of the Respiratory System

FURTHER READING

VetBooks.ir

Davis EG, Rush BR. Diagnostic challenges: equine thoracic neoplasia. Equine Vet Educ. 2013;25:96-107.

REFERENCES

1. Ortin A, et al. Vet Pathol. 2007;44:710. 2. Minguijon E, et al. J Comp Pathol. 2013;148:139. 3. Taintor J. Equine Vet Educ. 2014;26:499. 4. Sullins KE. Equine Vet Educ. 2015;27:306. 5. Van Heesewijk N, et al. Equine Vet Educ. 2015;27:302.

Congenital and Inherited Diseases of the Respiratory Tract CONGENITAL DEFECTS Primary congenital defects are rare in the respiratory tracts of animals. Congenital

defects of the soft palate of foals have been sporadically reported; horses with minor defects can grow normally and may be able to have a successful athletic career for their intended use.1 Hypoplasia of the epiglottis is detected occasionally in horses. Tracheal hypoplasia is recognized in calves and Miniature horses. Bronchogenic cysts are rare in foals2 and calves3 and result from the abnormal development of the tracheobronchial system during the embryonic period. Bronchogenic cysts can cause respiratory distress and dysphagia, particularly when located in the cervical region. Secondary defects, which are associated with major defects in other systems, are more common. Most of the defects are associated with defects of the oral cavity, face, and cranial vault, particularly cleft palate. Accessory lungs are recorded occasionally, and if their bronchi are vestigial, the lungs can present themselves as tumor-like masses occupying most of the

chest. Pulmonary hypoplasia has been associated with congenital diaphragmatic hernia. Retrosternal hernia (Morgagni hernia), which is a right ventral diaphragmatic defect, has been surgically corrected in adult horses as a result of incarceration of the large colon; in all cases the defect was thought to be congenital.4 REFERENCES

1. Barakzai SZ, et al. Equine Vet J. 2014;46:185. 2. Matsuda K, et al. Vet Pathol. 2010;47:351. 3. Lee JY, et al. J Vet Diagn Invest. 2010;22:479. 4. Pauwels FF, et al. J Am Vet Med Assoc. 2007;231:427.

VetBooks.ir

Index To access the comprehensive index for volumes 1 and 2 of VETERINARY MEDICINE, 11e please refer to the end of Volume 2.

1091

This page intentionally left blank

VetBooks.ir

VetBooks.ir

How to Use This Book We would like you to get the most out of this book. To do that, you should follow the directions provided in this section. And if you keep doing this every time you use the book, you will develop a proper diagnostic routine of going from: Clinical sign

System involved

Location and type of lesion

Specific cause of the disease

… and become what we wish for every one of you: a thinking clinician.

FOR EXAMPLE A yearling bull has a sudden onset of dyspnea, fever, anorexia, abnormal lung sounds, and nasal discharge. Step 1  The bull’s problem is dyspnea. Go to the index and find the principal entry for dyspnea. Step 2  The discussion on dyspnea will lead you to respiratory tract dyspnea and cardiac dyspnea. Step 3  Via the index, consult these and decide that the system involved is the respiratory system and that the lungs are the location of the lesion in the system. Step 4  Proceed to diseases of the lungs and decide on the basis of the clinical and other findings that the nature of the lesion is inflammatory and is pneumonia. Step 5  Proceed to pneumonia, and consult the list of pneumonias that occur in cattle. Consult each of them via the index and decide that pneumonic pasteurellosis is the probable specific cause. Step 6  Proceed to the section on pneumonic pasteurellosis and determine the appropriate treatment for the bull and the chances of saving it. Step 7  Don’t forget to turn to the end of the section on pneumonic pasteurellosis and remind yourself of what to do to protect the rest of the herd from sharing the illness.

Guidelines for Selection and Submission of Necropsy Specimens for Confirmation of Diagnosis In this edition we continue with the subheading Samples for Confirmation of Diagnosis to serve as a rough guideline for the collection of samples at necropsy. Several points must be emphasized with regard

to this section. First and foremost, collection of these samples is not advocated as a substitute for a thorough necropsy examination. Furthermore, the samples listed are selected to confirm the diagnosis, but a conscientious diagnostician should also collect samples that can be used to rule out other disease processes. Even the best of practitioners can make an incorrect tentative diagnosis, but it is an even more humbling experience if there are no samples available to pursue alternate diagnoses. Also, recall that some diseases may be the result of several different etiologic factors (e.g., neonatal diarrhea of calves), and the veterinarian who samples to confirm one of these factors but does not attempt to investigate others has not provided a good service to the client. A huge variety of veterinary diagnostic tests have been developed, but each veterinary diagnostic laboratory (VDL) offers only a selected panel, chosen after consideration of a number of factors. Such factors may include cost, demand, reliability, sensitivity and specificity, and the availability of appropriate technology at the lab. The array of diagnostic tests is constantly improving, and it is beyond the scope of this text to list all the tests available for a given disease or to recommend one test method to the exclusion of others. Under the Samples for Confirmation of Diagnosis sections, we have merely listed some of the more common tests offered. Advances in molecular biology are providing exciting avenues for disease diagnosis, but many of these tests have limited availability in VDLs at present. For optimal efficiency in the confirmation of a diagnosis at necropsy, the practitioner must contact the VDL to determine what tests are offered and to obtain the preferred protocol for sample collection and submission to that particular laboratory. Most VDLs publish user guidelines, which include the tests available and the samples required. The guidelines listed here are broad, and individual VDLs may have very specific requirements for sample handling. Several general statements can be made with regard to the submission of samples to VDLs: • The samples should be accompanied by a clearly written and concise clinical history, including the signalment of the animal and feeding and management information. Failure to provide this information deprives the owner of the full value of the expertise available from the laboratory staff. • If a potentially zoonotic disease is suspected, this should be clearly indicated in a prominent location on the submission form. • All specimens should be placed in an appropriate sealed, leak-proof container and clearly labeled with a waterproof marker to indicate the tissue/fluid collected, the animal sampled, and the owner’s name. At some VDLs, pooling of tissues within a single bag or container is permitted for specific tests (such as virus isolation), but in general, all fresh samples should be placed in separate containers. When packaging samples for shipment, recognize that condensation from ice packs and frozen tissues will damage any loose paper within the package; the submission sheet should be placed within a plastic bag for protection or taped to the outside of the shipping container. • Samples for histopathology can be pooled within the same container of 10% neutral-buffered formalin. An optimal tissue sample of a gross lesion should include the interface between normal and abnormal tissue. For proper fixation, tissue fragments should not be more than 0.5 cm in width, and the ratio of tissue to formalin solution should be 1 : 10. If necessary, large tissues such as brain can be fixed in a larger container and then transferred to a smaller one containing only a minimal quantity of formalin for shipping to the laboratory. To speed fixation and avoid artifactual changes, formalin containers should not be in direct contact with frozen materials during shipment.

IBC2

How to Use This Book

VetBooks.ir

• In the Samples for Confirmation of Diagnosis sections, the tests are listed under various discipline categories (bacteriology, virology, etc.). The appropriate sample(s) is noted, followed by the types of test that might be applied to these samples. The following is a list of these different tests, including any abbreviation used in this section of the text. A brief discussion of how the samples collected for each test should be handled is also provided. Again, it must be emphasized that this is by no means a complete listing of diagnostic tests available, and different VDLs often have differing sample handling procedures. • Aerobic culture = (CULT). These samples should generally be kept chilled during shipment. If a transit time of greater than 24 hours is anticipated the samples should be frozen, then packaged appropriately so that they are still frozen upon arrival at the VDL. Various bacterial species cannot be recovered using routine culture techniques, and most of these are highlighted in the text by the phrase “special culture requirements.” • Agar gel immunodiffusion = (AGID). A type of serologic test. Chilled or frozen serum may be submitted. • Anaerobic culture = (ANAEROBIC CULT). Confirmation of the diagnosis requires that any swabs be transported in special transport media and that the VDL attempts to grow bacteria from the samples under anaerobic culture conditions. Transport requirements are as for (CULT) (aerobic culture) specimens. • Analytical assay = (ASSAY). This refers to a broad range of tests in which a substance is quantitatively measured. The substance to be assayed is listed in brackets, e.g. (ASSAY [Ca]) denotes a test for calcium levels. The method used to perform the assay is not listed, but in general, frozen samples may be submitted for most of these analytical assays. • Bioassay = (BIOASSAY). This typically refers to tests in which the sample material is administered to an animal under experimental conditions. Preserved material is inappropriate, and some bioassays cannot be performed using samples that have been frozen. The VDL performing the test should be contacted for instructions prior to sample collection • Complement fixation = (CF). A serologic test. Ship chilled or frozen serum. • Cytology = (CYTO). Air-dried impression smears are usually adequate. Keep dry during transport. • Direct smear = (SMEAR). The type of test is usually given in brackets (e.g., [Gram]). Air-dried smears are usually adequate but must be kept dry during shipment. • Enzyme-linked immunosorbent assay = (ELISA). Chilled or frozen samples are usually acceptable. There are many variants of ELISA (e.g., antigen-capture, kinetic, indirect, direct, etc.), and the specific type used is not specified in this portion of the text. • Electron microscopic examination = (EM). Appropriate sample collection and handling varies with the specimen being examined. Most of the diagnostic specimens submitted to VDLs for EM are fecal samples, and these do not require any special preservative.

• Fecal floatation = (FECAL). Sample can be fresh, chilled, or frozen. • Fluorescent antibody test = (FAT). This may refer to either a direct or indirect method of antigen detection. Generally, cryostat sections are utilized, and therefore the tissue received by the laboratory should still be frozen upon arrival to provide the best results. Freeze/thaw cycles should be avoided. If impression smears are being shipped, they should be kept dry. • Fungal culture = (FCULT). Special media is required. Transport as per (CULT) specimens. • Immunohistochemical testing = (IHC). Many of these tests can be performed on formalin-fixed material, but in some instances frozen tissues must be delivered to the laboratory. In such instances the test is listed under a heading distinct from histology (e.g., virology, bacteriology, etc.). • Indirect hemagglutination = (IHA). A serologic test. Ship chilled or frozen serum. • In-situ hybridization = (IN-SITU HYBRID). Samples should be shipped chilled, although some test methods can use formalin-fixed material. These tests utilize nucleic acid probes that bind with complementary nucleic acid sequences in the specimen. Although not widely used in routine diagnostics at present, these methods may gain more prominence as their use is refined, • Virus isolation = (ISO). Samples should be kept chilled during shipment or maintained in a frozen state if prolonged transit times are anticipated, • Latex agglutination = (LATEX AGGLUTINATION). Fresh, chilled, or frozen samples are acceptable. • Light microscopic examination = (LM). Formalin-fixed tissues are preferred. The shipment of fresh tissues to the VDL permits more tissue autolysis prior to fixation, resulting in less useful specimens. If Bouin’s fixative is available, it is the preferred preservative for eye globes. • Microagglutination test = (MAT). A type of serologic test. Ship chilled or frozen serum. • Mycoplasmal culture = (MCULT). These types of organism have specific growth requirements that are usually not met by standard bacteriologic culture techniques. Transport as per (CULT) specimens. Culture swabs cannot be submitted in media containing charcoal or glycerol. • Polymerase chain reaction = (PCR). Tissues should be frozen and maintained in that state until arrival at the VDL. Swabs and fluids submitted for PCR testing should be chilled but not frozen. These tests are capable of detecting minute quantities of nucleic acid, so if multiple animals are tested, the samples should be “clean” to avoid false positives through cross-contamination (i.e., blood/tissue from one animal contaminating the sample from another) • Serum urea nitrogen = (SUN). A useful test to determine degree of renal compromise. Sample can be shipped chilled or frozen. • Virus neutralization = (VN). A serologic test. Ship chilled or frozen serum.

VetBooks.ir

VetBooks.ir

VETERINARY MEDICINE 11 EDITION

A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs, and Goats VOLUME TWO

PETER D. CONSTABLE KENNETH W. HINCHCLIFF STANLEY H. DONE WALTER GRÜNBERG

VetBooks.ir

3251 Riverport Lane St. Louis, Missouri 63043

VETERINARY MEDICINE: A TEXTBOOK OF THE DISEASES OF CATTLE, HORSES, SHEEP, PIGS, AND GOATS, ELEVENTH EDITION Copyright © 2017 Elsevier Ltd. All Rights Reserved. Previous editions copyrighted: 2007, 2000, 1999, 1994, 1983, 1979, 1974 First published 1960 No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions. This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

Notices Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary. Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility. With respect to any drug or pharmaceutical products identified, readers are advised to check the most current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the responsibility of practitioners, relying on their own experience and knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each individual patient, and to take all appropriate safety precautions. To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein. Main ISBN: 9780702052460 Volume 2 ISBN: 978-0-7020-7056-3

Content Strategist: Penny Rudolph Content Development Specialist: Laura Klein Content Development Manager: Jolynn Gower Publishing Services Manager: Hemamalini Rajendrababu Senior Project Manager: Kamatchi Madhavan Design Direction: Renee Duenow

Printed in China Last digit is the print number:  9  8  7  6  5  4  3  2  1

VetBooks.ir

Contents Contributors,  ix Preface to the Eleventh Edition,  x Introduction,  xii List of Tables,  xxiv List of Illustrations,  xxvii

1 Clinical Examination and Making a Diagnosis,  1 Introduction,  1 Making a Diagnosis,  2 Clinical Examination of the Individual Animal,  5 Prognosis and Therapeutic Decision Making,  26

2 Examination of the Population,  29 Approach to Examining the Population,  29 Examination Steps,  30 Techniques in Examination of the Herd or Flock,  32 Role of the Integrated Animal Health and Production Management Program,  34

3 Biosecurity and Infection Control,  36 Definitions and Concepts,  36 Development of a Biosecurity   Plan,  37 Practices to Aid in Maintaining Biosecurity,  38

4 General Systemic States,  43 Hypothermia, Hyperthermia, and Fever,  43 Acute Phase Response,  56 Sepsis, Septicemia, and Viremia,  57 Toxemia, Endotoxemia, and Septic Shock,  59 Toxemia in the Recently Calved Cow,  67 Hypovolemic, Hemorrhagic, Maldistributive, and Obstructive Shock,  71 Localized Infections,  76 Pain,  78 Stress,  84 Disturbances of Appetite, Food Intake, and Nutritional Status,  87 Weight Loss or Failure to Gain Weight (Ill-Thrift),  90 Physical Exercise and Associated Disorders,  96 Sudden or Unexpected Death,  99 Diseases Associated With Physical Agents,  103 Diagnosis of Inherited Disease,  111

5 Disturbances of Free Water, Electrolytes, Acid-Base Balance, and Oncotic Pressure,  113 Dehydration,  113 Water Intoxication,  115 Electrolyte Imbalances,  116 Acid-Base Imbalance,  123 Oncotic Pressure and Edema,  128 Naturally Occurring Combined Abnormalities of Free Water, Electrolyte, Acid-Base Balance, and Oncotic Pressure,  130 Principles of Fluid and Electrolyte Therapy,  137

6 Practical Antimicrobial Therapeutics,  153 Principles of Antimicrobial   Therapy,  153 Antibiotic Resistance,  156 Antibiotic Metaphylaxis to Control Respiratory Disease,  158 Practical Usage of Antimicrobial Drugs,  158 Classification of Antimicrobial Agents: Mechanisms of Action and Major Side Effects,  169 β-Lactam Antibiotics: Penicillins, Cephalosporins, and β-Lactamase Inhibitors,  170

7 Diseases of the Alimentary Tract: Nonruminant,  175 Principles of Alimentary Tract Dysfunction,  176 Manifestations of Alimentary Tract Dysfunction,  178 Special Examination,  183 Principles of Treatment in Alimentary Tract Disease,  190 Diseases of the Buccal Cavity and Associated Organs,  192 Diseases of the Pharynx and Esophagus,  196 Diseases of the Nonruminant Stomach and Intestines,  203 Diseases of the Peritoneum,  215 Abdominal Diseases of the Horse Including Colic and Diarrhea,  220 Abdominal Diseases of the Pig Including Diarrhea,  287 Noninfectious Intestinal Disease of Swine,  290 Bacterial and Viral Diseases of the Alimentary Tract,  292 Parasitic Diseases of the Alimentary Tract,  397 Control,  421 Toxins Affecting the Alimentary Tract,  421 Neoplasms of the Alimentary Tract,  431

Congenital Defects of the Alimentary Tract,  432 Inherited Defects of the Alimentary Tract,  434

8 Diseases of the Alimentary Tract–Ruminant,  436 Diseases of the Forestomach of Ruminants,  436 Special Examination of the Alimentary Tract and Abdomen of Cattle,  445 Diseases of the Rumen, Reticulum and Omasum,  457 Diseases of the Abomasum,  500 Diseases of the Intestines of Ruminants,  523 Bacterial Diseases of the Ruminant Alimentary Tract,  531 Viral Diseases of the Ruminant Alimentary Tract,  572 Parasitic Diseases of the Ruminant Alimentary Tract,  603 Toxic Diseases of the Ruminant Alimentary Tract,  618 Diseases of the Ruminant Alimentary Tract of Unknown Cause,  621

9 Diseases of the Liver,  622 Diseases of the Liver: Introduction,  622 Principles of Hepatic Dysfunction,  622 Manifestations of Liver and Biliary Disease,  623 Special Examination of the Liver,  625 Principles of Treatment in Diseases of the Liver,  629 Diffuse Diseases of the Liver,  629 Diseases Characterized by Systemic Involvement,  639 Hepatic Diseases Associated With Trematodes,  641 Diseases Associated With Major Phytotoxins,  645 Poisoning by Mycotoxins,  649 Focal Diseases of the Liver,  655 Diseases of the Pancreas,  656

10 Diseases of the Cardiovascular System,  657 Principles of Circulatory Failure,  657 Manifestations of Circulatory Failure,  659 Special Examination of the Cardiovascular System,  663 Arrhythmias (Dysrhythmias),  675 Diseases of the Heart,  685 Cardiac Toxicities,  697 Cardiac Neoplasia,  703 Congenital Cardiovascular Defects,  703 Inherited Defects of the Circulatory System,  706 Diseases of the Pericardium,  707 Diseases of the Blood Vessels,  709 Vascular Neoplasia,  715 iii

iv

Contents

VetBooks.ir

11 Diseases of the Hemolymphatic and Immune Systems,  716 Abnormalities of Plasma Protein Concentration,  716 Hemorrhagic Disease,  718 Lymphadenopathy (Lymphadenitis),  751 Diseases of the Spleen and Thymus,  752 Immune-Deficiency Disorders (Lowered Resistance to Infection),  753 Amyloidoses,  755 Enzootic Bovine Leukosis (Bovine Lymphosarcoma),  785 Nutritional Deficiencies,  814 Toxins Affecting the Hemolymphatic System,  823 Neoplasia,  834 Congenital Inherited Diseases,  837 Inherited Immunodeficiency,  839 Diseases of Unknown Etiology,  842

12 Diseases of the Respiratory System,  845 Principles of Respiratory Insufficiency,  846 Principal Manifestations of Respiratory Insufficiency,  848 Special Examination of the Respiratory System,  855 Principles of Treatment and Control of Respiratory Tract Disease,  868 Diseases of the Upper Respiratory Tract,  874 Diseases of the Lung   Parenchyma,  880 Diseases of the Pleural Cavity and Diaphragm,  895 Diseases of the Bovine Respiratory Tract,  901 Diseases of the Ovine and Caprine Respiratory Tract,  969 Diseases of the Equine Respiratory Tract,  981 Diseases of the Swine Respiratory Tract,  1047 Respiratory System Toxicoses,  1087 Neoplastic Diseases of the Respiratory Tract,  1088 Congenital and Inherited Diseases of the Respiratory Tract,  1090

13 Diseases of the Urinary System,  1095 Introduction,  1095 Clinical Features of Urinary Tract Disease,  1097 Special Examination of the Urinary System,  1099 Principles of Treatment of Urinary Tract Disease,  1108 Diseases of the Kidney,  1110 Infectious Diseases of   the Kidney,  1115 Toxic Agents Affecting   the Kidney,  1135 Renal Neoplasia,  1137 Congenital and Inherited Renal Diseases,  1137

Diseases of the Ureters, Bladder, and Urethra,  1139 Diseases of the Prepuce and Vulvovaginal Area,  1152

14 Diseases of the Nervous System,  1155 Introduction,  1156 Principles of Nervous   Dysfunction,  1157 Clinical Manifestations of Diseases of the Nervous System,  1158 Special Examination of the Nervous System,  1164 Diffuse or Multifocal Diseases of the Brain and Spinal Cord,  1178 Focal Diseases of the Brain and Spinal Cord,  1189 Plant Toxins Affecting the Nervous System,  1194 Fungal Toxins Affecting the Nervous System,  1201 Other Toxins Affecting the Nervous System,  1202 Diseases of the Cerebrum,  1219 Bacterial Diseases Primarily Affecting the Cerebrum,  1224 Viral Diseases Primarily Affecting the Cerebrum,  1227 Prion Diseases Primarily Affecting the Cerebrum,  1286 Parasitic Disease Primarily Affecting the Cerebrum,  1301 Metabolic Diseases Primarily Affecting the Cerebrum,  1302 Metabolic and Toxic Encephalomyelopathies,  1321 Inherited Diseases Primarily Affecting the Cerebrum,  1322 Congenital and Inherited Encephalomyelopathies,  1324 Diseases Primarily Affecting the Cerebellum,  1328 Diseases Primarily Affecting the Brainstem and Vestibular System,  1329 Diseases Primarily Affecting the Spinal Cord,  1337 Parasitic Diseases Primarily Affecting the Spinal Cord,  1341 Toxic Diseases Primarily Affecting the Spinal Cord,  1346 Inherited Diseases Primarily Affecting the Spinal Cord,  1346 Diseases Primarily Affecting the Peripheral Nervous System,  1358

15 Diseases of the Musculoskeletal System,  1371 Principal Manifestations of Musculoskeletal Disease,  1372 Diseases of Muscles,  1377 Diseases of Bones,  1388 Diseases of Joints,  1406 Infectious Diseases of the Musculoskeletal System,  1425 Nutritional Diseases Affecting the Musculoskeletal System,  1458

Toxic Agents Affecting the Musculoskeletal System,  1503 Congenital Defects of Muscles, Bones, and Joints,  1510 Inherited Diseases of Muscles,  1514 Inherited Diseases of Bones,  1530 Inherited Diseases of Joints,  1538

16 Diseases of the Skin, Eye, Conjunctiva, and External Ear,  1540 Introduction,  1541 Principles of Treatment of Diseases of the Skin,  1543 Diseases of the Epidermis and Dermis,  1543 Diseases of the Hair, Wool, Follicles, and Skin Glands,  1552 Diseases of the Subcutis,  1555 Non-Infectious Diseases of the Skin,  1559 Bacterial Diseases of the Skin,  1564 Viral Diseases of the Skin,  1580 Dermatomycoses,  1600 Protozoal Diseases of the Skin,  1607 Nematode Infections of   the Skin,  1608 Cutaneous Myiasis,  1611 Mite Infestations,  1618 Ked and Louse Infestations,  1623 Miscellaneous Skin Diseases Caused   by Flies, Midges, and Mosquitoes,  1625 Tick Infestations,  1631 Deficiencies and Toxicities Affecting the Skin,  1634 Cutaneous Neoplasms,  1640 Congenital and Inherited Defects of the Skin,  1643 Eye and Conjunctival Diseases,  1648 External Ear Diseases,  1660

17 Metabolic and Endocrine Diseases,  1662 Introduction,  1662 Metabolic Diseases of   Ruminants,  1662 Inherited Metabolic Diseases of Ruminants,  1727 Metabolic Diseases of Horses,  1727 Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma),  1739 Diseases Caused by Nutritional Deficiencies,  1747 Deficiencies of Energy and Protein,  1753 Diseases Associated with Deficiencies of Mineral Nutrients,  1754

18 Diseases Primarily Affecting the Reproductive System,  1758 Infectious Diseases Primarily   Affecting the Reproductive System,  1758 Infectious Diseases Primarily Affecting the Reproductive System,  1761

Contents

VetBooks.ir

Toxic Agents Primarily Affecting the Reproductive System,  1821 Congenital and Inherited Diseases Primarily Affecting the Reproductive System,  1828

19 Perinatal Diseases,  1830 Introduction,  1830 Perinatal and Postnatal Diseases,  1830 Perinatal Disease—Congenital Defects,  1835 Physical and Environmental Causes of Perinatal Disease,  1840 Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin),  1848 Clinical Assessment and Care of Critically Ill Newborns,  1856 Neonatal Infectious Diseases,  1874 Neonatal Neoplasia,  1903

20 Diseases of the Mammary Gland,  1904 Introduction,  1904 Bovine Mastitis,  1904 Diagnosis of Bovine Mastitis,  1914 Mastitis Pathogens of Cattle,  1930

Mastitis of Cattle Associated With Common Contagious Pathogens,  1930 Mastitis of Cattle Associated With Teat Skin Opportunistic Pathogens,  1942 Mastitis of Cattle Associated With Common Environmental Pathogens,  1943 Mastitis of Cattle Associated With Less Common Pathogens,  1960 Control of Bovine Mastitis,  1964 Miscellaneous Abnormalities of the Teats and Udder,  1985 Mastitis of Sheep,  1991 Mastitis of Goats,  1993 Contagious Agalactia in Goats and Sheep,  1994 Mastitis of Mares,  1996 Postpartum Dysgalactia Syndrome of Sows,  1996

21 Systemic and Multi-Organ Diseases,  2002 Diseases of Complex or Undetermined Etiology,  2003 Multi-Organ Diseases Due to Bacterial Infection,  2011

v

Multi-Organ Diseases Due to Viral Infection,  2058 Multi-Organ Diseases Due to Protozoal Infection,  2137 Multi-Organ Diseases Due to Trypanosome Infection,  2150 Multi-Organ Diseases Due to Fungal Infection,  2158 Multi-Organ Diseases Due to Metabolic Deficiency,  2161 Multi-Organ Diseases Due to Toxicity,  2176

APPENDICES,  2215 Appendix 1  Conversion Tables,  2215 Appendix 2   Reference Laboratory Values,  2217 Appendix 3  Drug doses and intervals for horses and ruminants,  2220 Appendix 4  Drug doses and intervals for pigs,  2232

Index,  2235

This page intentionally left blank

VetBooks.ir

VetBooks.ir

Diseases of the Urinary System INTRODUCTION  1095 Principles of Renal Insufficiency  1095 Renal Insufficiency and Renal Failure  1096 CLINICAL FEATURES OF URINARY TRACT DISEASE  1097 Abnormal Constituents of the Urine  1097 Variations in Daily Urine Flow  1097 Abdominal Pain and Painful and Difficult Urination (Dysuria and Stranguria)  1098 Morphologic Abnormalities of Kidneys and Ureters  1098 Palpable Abnormalities of the Bladder and Urethra  1098 Acute and Chronic Renal Failure  1098 Uremia  1098 SPECIAL EXAMINATION OF THE URINARY SYSTEM  1099 Tests of Renal Function and Detection of Renal Injury  1099 Collection of Urine Samples  1099 Tests of Urine Samples  1100 Tests of Serum  1103 Tests of Urine and Serum  1104 Diagnostic Examination Techniques  1106 PRINCIPLES OF TREATMENT OF URINARY TRACT DISEASE  1108 DISEASES OF THE KIDNEY  1110 Glomerulonephritis  1110

Introduction Diseases of the bladder and urethra are more common and more important than diseases of the kidneys in farm animals. Occasionally, renal insufficiency develops as a sequel to diseases such as pyelonephritis, embolic nephritis, amyloidosis, and nephro­ sis. Knowledge of the physiology of urinary secretion and excretion is required to prop­ erly understand disease processes in the urinary tract. The principles of renal insuf­ ficiency presented here are primarily extra­ polated from research in other species, particularly human medicine. Although gen­ erally these principles probably apply to farm animals, the details of renal function and renal failure in farm animals have just started to be studied in depth.

13 

Pyelonephritis  1111 Nephrosis  1111 Ischemic Nephrosis  1112 Toxic Nephrosis  1112 Renal Tubular Acidosis  1113 Hemolytic Uremic–Like Syndrome  1114 Hydronephrosis  1114 Interstitial Nephritis  1114 Embolic Nephritis  1115 INFECTIOUS DISEASES OF THE KIDNEY  1115 Leptospirosis  1115 Bovine Pyelonephritis  1129 Urinary Disease in Swine  1131 Porcine Cystitis and Pyelonephritis  1132 Kidney Worm Disease in Pigs Caused by Stephanurus dentatus  1134 TOXIC AGENTS AFFECTING THE KIDNEY  1135 Citrinin Toxicosis  1135 Ethylene Glycol Toxicosis  1136 Ochratoxins (Ochratoxicosis)  1136 Plant Poisonings Caused by Known Toxins  1137 Plant Poisonings From Unidentified Toxins  1137 Fungi Lacking Identified Toxins  1137 RENAL NEOPLASIA  1137

CONGENITAL AND INHERITED DISEASES OF THE KIDNEY  1137 Renal Hypoplasia  1137 Polycystic Kidneys  1138 Renal Dysplasia  1138 Renal Lipofucinosis of Cattle  1139 Equine Renal Cortical Tubular Ectasia  1139 DISEASES OF THE URETERS, BLADDER, AND URETHRA  1139 Ectopic Ureter and Ureteral Defects  1139 Paralysis of the Bladder and Overflow Incontinence  1139 Eversion of the Bladder  1140 Patent Urachus  1140 Rupture of the Bladder (Uroperitoneum)  1140 Uroperitoneum in Foals  1140 Cystitis  1143 Urolithiasis in Ruminants  1144 Urolithiasis in Horses  1150 Urethral Tears in Stallions and Geldings  1151 Urethral Defects  1151 Urinary Bladder Neoplasms  1151 Bovine Enzootic Hematuria  1151 DISEASES OF THE PREPUCE AND VULVOVAGINAL AREA  1152 Enzootic Posthitis (Pizzle Rot, Sheath Rot, Balanoposthitis) and Vulvovaginitis (Scabby Ulcer)  1152

PRINCIPLES OF RENAL INSUFFICIENCY

passage of high molecular weight sub­ stances, such as plasma proteins. Glomeru­ lar filtrate is derived from plasma by simple passive filtration driven by arterial blood pressure. Glomerular filtrate is identical to plasma except that it contains little protein or lipids. The volume of filtrate, and there­ fore its content of metabolic end products, depends on the hydrostatic pressure and the plasma oncotic pressure in the glomerular capillaries and on the proportion of glom­ eruli, which are functional. Because these factors are only partially controlled by the kidney, in the absence of disease, the rate of filtration through the glomeruli is relatively constant. Epithelial cells in the renal tubules actively and selectively reabsorb substances from the glomerular filtrate while permitting the excretion of waste products. Proximal

The kidneys excrete the end products of tissue metabolism (except for carbon dioxide), and maintain fluid, electrolyte, and acid-base balance, by varying the volume of water and the concentration of solutes in the urine. For conceptual purposes it is helpful to think of the kidney as composed of many similar nephrons, which are the basic func­ tional units of the kidney. Each nephron is composed of blood vessels, the glomerulus, and a tubular system that consists of the proximal tubule, the loop of Henle, the distal tubule, and the collecting duct. The glomerulus is a semipermeable filter that allows easy passage of water and low molecular weight solutes, such as electro­ lytes, glucose, and keto acids, but restricts

Copyright © 2017 Elsevier Ltd. All Rights Reserved. 1095

1096

Chapter 13 

■ 

Diseases of the Urinary System

VetBooks.ir

tubular cells are therefore metabolically very active and, consequently, are susceptible to injury from ischemia (decreased blood blood) or hypoxia. Glucose is reabsorbed entirely, within the normal range of plasma concentration; phosphate is reabsorbed in varying amounts depending on the needs of the body for phosphorus conservation; other substances, such as inorganic sulfates and creatinine, are not reabsorbed in appreciable amounts. The tubules also actively secrete substances, particularly electrolytes, as they function to regulate acid-base balance. As a result of the balance between resorption and secretion, the concentration of solutes in the urine varies widely when the kidneys are functioning normally. The principal mechanism that regulates water reabsorption by the renal tubules is antidiuretic hormone (ADH). Tissue dehy­ dration and an increase in serum osmolality stimulate the secretion of ADH from the posterior pituitary gland. The renal tubules respond to ADH by conserving water and returning serum osmolality to normal, pro­ ducing concentrated urine. Diseases of the kidneys, and in some instances of the ureters, bladder, and urethra, reduce the efficiency of the kidney’s func­ tions, resulting in disturbances in protein, acid-base, electrolyte, and water homeostasis and in the excretion of metabolic end prod­ ucts. A partial loss of function is described as renal insufficiency. When the kidneys can no longer regulate body fluid and solute composition, renal failure occurs.

RENAL INSUFFICIENCY AND RENAL FAILURE Renal function depends on the number and functionality of the individual nephrons. Renal insufficiency can occur from abnor­ malities in the • Rate of renal blood flow • Glomerular filtration rate • Efficiency of tubular reabsorption. Of these three abnormalities, the latter two are intrinsic functions of the kidney, whereas the first depends largely on vasomo­ tor control, which is markedly affected by circulatory emergencies such as shock, dehy­ dration, and hemorrhage. Circulatory emer­ gencies may lead to a marked reduction in glomerular filtration, but they are extrarenal in origin and cannot be considered as true causes of renal insufficiency. However, pro­ longed circulatory disruption can cause renal ischemia and ultimately renal insufficiency. Glomerular filtration and tubular reab­ sorption can be affected independently in disease states, and every attempt should be made to clinically differentiate glomerular disease from tubular disease. This is because the clinical and clinicopathologic signs of renal dysfunction depend on the anatomic location of the lesion and the imbalance in function between glomeruli and tubules.

Renal dysfunction tends to be a dynamic process so the degree of dysfunction varies with time. If renal dysfunction is so severe that the animal’s continued existence is not possible, it is said to be in a state of renal failure, and the clinical syndrome of uremia will be present.

CAUSES OF RENAL INSUFFICIENCY AND UREMIA

The causes of renal insufficiency, and there­ fore of renal failure and uremia, can be divided into prerenal, renal, and postrenal groups. Prerenal causes include congestive heart failure and acute circulatory failure, either cardiac or peripheral, in which acute renal ischemia occurs in response to a decrease in renal blood flow. Proximal tubular function is affected by renal ischemia to a much greater extent than the glomerulus or distal tubules; this is because of the high metabolic demands of the proximal tubules. However, those parts of the tubules within the medulla are particu­ larly susceptible to hypoxic damage because of the low oxygen tension in this tissue, the dependency of blood flow on glomerular blood flow, and the high metabolic rate of this tissue. Renal medullary necrosis is a direct consequence of these factors. Renal causes include glomerulonephri­ tis, amyloidosis, pyelonephritis, embolic nephritis, and interstitial nephritis. Acute renal failure can be produced in any of the farm animal species by administration of a variety of toxins (see the section Toxic Nephrosis). The disease is also secondary to sepsis and hemorrhagic shock. Experimental uremia has also been induced by surgical removal of both kidneys but the results, especially in ruminants, are quite different from those in naturally occurring renal failure. The clinical pathology is similar, but there is a prolonged period of normality after the surgery. Postrenal uremia may also occur, spe­ cifically complete obstruction of the urinary tract by vesical or urethral calculus, or more rarely by bilateral urethral obstruction by transitional cell carcinoma located in the trigone region of the bladder. Internal rupture of any part of the urinary tract, such as the bladder, ureters, or urethra, will also cause postrenal uremia.

PATHOGENESIS OF RENAL INSUFFICIENCY AND RENAL FAILURE

Damage to the glomerular epithelium destroys its selective permeability and per­ mits the passage of plasma proteins into the glomerular filtrate. The predominant protein is initially albumin, because of its negative charge and a lower molecular weight than globulins; however, with advanced glomeru­ lonephritis (such as renal amyloidosis) all plasma proteins are lost. Glomerular filtra­ tion may cease completely when there is

extensive damage to glomeruli, particularly if there is acute swelling of the kidney, but it is thought that anuria in the terminal stages of acute renal disease is caused by back dif­ fusion of all glomerular filtrate through the damaged tubular epithelium rather than fail­ ure of filtration. When renal damage is less severe, the remaining nephrons compensate to maintain total glomerular filtration by increasing their filtration rates. When this occurs, the volume of glomerular filtrate may exceed the capacity of the tubular epithelium to reabsorb fluid and solutes. The tubules may be unable to achieve normal urine con­ centration. As a result, an increased volume of urine with a constant specific gravity is produced and solute diuresis occurs. This is exacerbated if the tubular function of the compensating nephrons is also impaired. The inability to concentrate urine is clinically evident as polyuria and is characteristic of developing renal insufficiency. Decreased glomerular filtration also results in retention of metabolic waste prod­ ucts such as urea and creatinine. Although marked increases in serum urea concentra­ tion are probably not responsible for the production of clinical signs, because urea readily crosses cell membranes and therefore is an ineffective osmole, the serum urea nitrogen (SUN) concentration can be used to monitor glomerular filtration rate. How­ ever, the utility of SUN concentration as a measure of glomerular filtration rate is reduced because serum urea concentrations are influenced by the amount of protein in the diet, by hydration, and by gastrointestinal metabolism of urea. Serum urea concentra­ tions are substantially higher in animals on high-protein diets, and dehydration increases serum urea concentration by increasing resorption of urea in the loop of Henle, which is independent of effects of hydration of the glomerular filtration rate. Urea is excreted into saliva of ruminants and metabolized by ruminal bacteria. In contrast, creatinine is excreted almost entirely by the kidney, creatinine originates from the breakdown of creatine phosphate in muscle, and serum concentrations of creatinine are a useful marker of glomerular filtration rate. The relationship between serum creatinine con­ centration and glomerular filtration rate is hyperbolic (a reduction in glomerular filtra­ tion rate by half results in a doubling of the serum creatinine concentration). Phosphate and sulfate retention also occurs when total glomerular filtration is reduced and sulfate retention contributes to metabolic acidosis in renal insufficiency. Phosphate retention also causes a secondary hypocalcemia, due in part to an increase in calcium excretion in the urine. In horses, the kidneys are an important route of calcium excretion; thus the decreased glomerular filtration rate present in horses with chronic renal failure usually results in hypercalcemia. Variations in serum potas­ sium concentrations also occur and appear to

Clinical Features of Urinary Tract Disease

VetBooks.ir

depend on potassium intake. Hyperkalemia is not usually a serious complication of renal insufficiency in ruminants because affected animals often have decreased appetites and therefore reduced potassium intakes, and excess saliva can be excreted by the salivary glands and ultimately the feces. Loss of tubular resorptive function is evi­ denced by a continued loss of sodium and chloride; hyponatremia and hypochloremia eventually occur in all cases of renal failure. The continuous loss of large quantities of fluid from solute diuresis can cause clinical dehydration. More often it makes the animal particularly susceptible to dehydration when there is an interruption in water availability or when there is a sudden increase in body water loss by another route, as in diarrhea. The terminal stage of renal insufficiency, renal failure, is the result of the cumulative effects of impaired renal excretory and homeostatic functions. Sustained excretion of large volumes of dilute urine results in dehydration. If other circulatory emergen­ cies arise, acute renal ischemia might result, leading to acute renal failure. Prolonged hypoproteinemia results in rapid loss of body condition and muscle weakness. Acidemia secondary to metabolic acidosis and hypona­ tremia can also be a contributing factor to muscle weakness and mental attitude. All these factors play some part in the produc­ tion of clinical signs of renal failure, which are typically manifested as weakness, leth­ argy, inappetence and, with extensive glo­ merular lesions, dependent edema caused by hypoproteinemia. However, the clinical syn­ drome is variable and rarely diagnostic for renal failure. Bleeding diathesis can also be present in severely uremic animals and has been associated with a lack of antithrombin (a small protein readily lost through the damaged glomerulus), platelet factor 3, platelet dysfunction, or disseminated intra­ vascular coagulation. Renal failure is seen as the clinical state of uremia. Uremic animals exhibit clinical signs of disease, which should be compared with azotemic animals that have an increase in the plasma or serum concentrations of urea and creatinine and retention of other solutes as described earlier, but do not necessarily have clinical signs of disease.

Clinical Features of Urinary Tract Disease The major clinical manifestations of urinary tract disease are • Abnormal constituents of urine • Variations in daily urine flow • Abdominal pain, painful urination (dysuria), and difficult urination (dysuria and stranguria) • Abnormal sized kidneys

• Abnormalities of the bladder and urethra • Acute and chronic renal failure

ABNORMAL CONSTITUENTS OF THE URINE Laboratory analysis of urine is initially done using dipstick and refractometry on a voided or catheterized urine sample and microscopic examination of the sediment from a centri­ fuged urine sample. Urine dipsticks and refractometry (optical and digital) provide excellent low-cost point-of-care tests for eval­ uation of the urinary system. Widely available urine dipsticks typically measure 1 factor (acetoacetate), 5 factors (blood, glucose, ace­ toacetate, pH, and protein) or 10 factors (blood, glucose, bilirubin, acetoacetate, pH, protein, specific gravity, urobilinogen, nitrite, and leukocytes). Specific information regard­ ing tests of renal function and injury con­ ducted on urine, such as specific gravity and osmolality, enzymuria, and quantitative pro­ teinuria and glycosuria, are discussed later in this chapter.

VARIATIONS IN DAILY URINE FLOW An increase or decrease in urine flow is often described in animals, but accuracy demands physical measurement of the amount of urine voided over a 24-hour period. This is not usually practicable in large-animal prac­ tice, and it is often necessary to guess whether the flow is increased or decreased. Accurate measurement of the amount of water con­ sumed is often easier and is usually used to estimate 24-hour urine production. Care should be taken to differentiate increased daily urine flow from increased frequency in urination without increased daily flow. The latter is much more common. Decreased urine output rarely, if ever, presents as a clini­ cal problem in agricultural animals. Normal urine production is highly vari­ able in large animals and is dependent to a large extent on diet, watering systems, and the palatability of the water. Pregnant mares housed in tie stalls consume approximately 53 ± 6 mL of water per kilogram body weight (BW) per day, of which 50 ± 8 mL/kg is from drinking water with the remainder being water in feed. However, most of this water is excreted in the feces, with fecal and urinary water excretion being 34 ± 8 (mL/kg)/day and 8 ± 2 (mL/kg)/day, respectively. Neona­ tal foals produce urine at an average rate of 150 (mL/kg)/day. Polyuria Polyuria occurs when there is an increase in the volume of urine produced over a 24-hour period. Polyuria can result from extrarenal causes, such as when horses habitually drink excessive quantities of water (psychogenic polydipsia) and, much less common, in

1097

central diabetes insipidus, when there is inappropriate secretion of ADH from the pituitary, or when there is failure of the tubules to respond to ADH (nephrogenic diabetes insipidus). Polyuria occurs in horses with tumors of the pars intermedia of the pituitary gland. Although the cause of the polyuria is not known, it might be secondary to osmotic diuresis associated with the glucosuria or to central diabetes insipidus. Central diabetes insipidus is reported in sibling colts but is extremely rare in other species with isolated reports in a ram and a cow. Another extrarenal cause is administration of diuretic drugs, including corticosteroids. Kidney disease results in polyuria when the resorptive capacity of the remaining tubules is exceeded. Polyuria can also occur when the osmotic gradient in the renal medulla is not adequate to produce concen­ trated urine. Nephrogenic diabetes insipidus causes polyuria because the tubules fail to respond to ADH. When polyuria is suspected, a urine sample should be collected to determine spe­ cific gravity or osmolality. If urine is isosthe­ nuric with a constant specific gravity of 1.008 to 1.012 (the specific gravity of plasma), then the presence of renal disease should be strongly considered. Serum urea and creati­ nine concentrations should be determined to evaluate glomerular filtration. If serum urea and creatinine concentrations are within normal limits, a water deprivation test can be performed to assess the animal’s ability to produce concentrated urine. Oliguria and Anuria Reduction in the daily output of urine (oliguria) and complete absence of urine (anuria) occur under the same conditions and vary only in degree. In dehydrated animals, urine flow naturally decreases in an effort to conserve water as plasma osmo­ lality increases. Congestive heart failure and peripheral circulatory failure may cause a reduction in renal blood flow that oliguria follows. Complete anuria is most common in urethral obstruction, although it can also result from acute tubular nephrosis. Oli­ guria occurs in the terminal stages of all forms of nephritis. Anuria and polyuria lead to retention of solutes and disturbances of the acid-base balance that contribute to the pathogenesis of uremia. Pollakiuria This is an increase in the daily number of postures for urination and is usually accom­ panied by a decreased volume of urine. Pol­ lakiuria may occur with or without an increase in the volume of urine excreted and is commonly associated with disease of the lower urinary tract such as cystitis, the presence of calculi in the bladder, ure­ thritis, and partial obstruction of the urethra. Other causes of pollakiuria include equine

1098

Chapter 13 

■ 

Diseases of the Urinary System

VetBooks.ir

herpesvirus infection, sorghum cystitis, neu­ ritis of the cauda equina in horses, neoplasia, obstructive lesions and trauma to the urethra, abnormal vaginal conformation, and urachal infection. Dribbling is a steady, intermittent passage of small volumes of urine, some­ times precipitated by a change in posture or increase in intraabdominal pressure, reflect­ ing inadequate or lack of sphincter control. Dribbling occurs in large animals with incomplete obstructive urolithiasis and from persistent urachus. Persistent urachus is also called pervious or patent urachus. Failure of the urachus to obliterate at birth in foals causes urine to dribble from the urachus continuously. Urine may also pass from the urethra. Retro­ grade infection from omphalitis is common, resulting in cystitis. Persistent urachus is extremely rare in calves, lambs, and kids. Abnormalities of micturition are classi­ fied as neurogenic or nonneurogenic. Mictu­ rition is mediated principally by the pelvic and pudendal nerves through lumbosacral spinal cord segments under the involuntary control of centers in the brainstem and vol­ untary control of the cerebrum and cerebel­ lum. Reported neurogenic causes of urinary incontinence in horses include cauda equine neuritis, herpesvirus-1 myelitis, Sudan grass toxicosis, sorghum poisoning, trauma, and neoplasia. Nonneurogenic causes of urinary incontinence in horses include ectopic ureter, cystitis, urolithiasis, hypoestrogen­ ism, and abnormal vaginal conformation.

ABDOMINAL PAIN AND PAINFUL AND DIFFICULT URINATION (DYSURIA AND STRANGURIA) Abdominal pain and painful urination (dysuria) and difficult and slow urination (stranguria) are manifestations of discom­ fort caused by disease of the urinary tract. Acute abdominal pain from urinary tract disease occurs only rarely and is usually associated with sudden distension of the renal pelvis or ureter, or infarction of the kidney. None of these conditions is common in animals, but occasionally cattle affected with pyelonephritis may have short episodes of acute abdominal pain caused by either renal infarction or obstruction of the pelvis by necrotic debris. During these acute attacks of pain, the cow may exhibit downward arching of the back, paddling with the hind feet, rolling, and bellowing. Abdominal pain from urethral obstruction and distension of the bladder is manifested by tail switching, kicking at the belly, and repeated straining efforts at urination accompanied by grunt­ ing. Horses with acute tubular nephrosis fol­ lowing vitamin K3 administration might show renal colic with arching of the back, backing into corners, and rubbing of the perineum and tail head.

Dysuria or painful/difficult urination occurs in cystitis, vesical calculus, urethritis, and is caused by the presence of periurethral masses such as pelvic lymphoma.1 Dysuria is manifested by the frequent passage of small amounts of urine. Grunting may occur with painful urination, and the animal may remain in the typical posture after urination is com­ pleted. Differentiating pain caused by urinary disease from pain caused by other causes depends largely on the presence of other signs indicating urinary tract involvement. Stranguria is slow and painful urination associated with disease of the lower urinary tract including cystitis, vesical calculus, ure­ thral obstruction, and urethritis. The animal strains to pass each drop of urine. Groaning and straining may precede and accompany urination when there is urethral obstruction. In urethritis, groaning and straining occur immediately after urination has ceased and gradually disappear and do not recur until urination has been repeated. Urine scalding of the perineum or urinary burn is caused by frequent wetting of the skin with urine. It may be the result of urinary incontinence or the animal’s inability to assume normal posture when urinating.

MORPHOLOGIC ABNORMALITIES OF KIDNEYS AND URETERS Enlarged or decreased size of kidneys may be palpable on rectal examination or detected by ultrasonography. In cattle, gross enlargement of the posterior aspect of the left kidney may be palpable in the right upper flank. Abnor­ malities of the kidneys, such as hydronephro­ sis in cattle, may also be palpable on rectal examination. Increases in the size of the ureter may be palpable on rectal examination and indicate ureteritis or hydroureter.

PALPABLE ABNORMALITIES OF THE BLADDER AND URETHRA Abnormalities of the bladder that may be palpable by rectal examination include gross enlargement of the bladder, rupture of the bladder, a shrunken bladder following rup­ ture, and palpable abnormalities in the blad­ der such as cystic calculi. Abnormalities of the urethra include enlargement and pain of the pelvic urethra and its external aspects in male cattle with obstructive urolithiasis and obstruction of the urethral process of male sheep with obstructive urolithiasis.

ACUTE AND CHRONIC RENAL FAILURE The clinical findings of urinary tract disease vary with the rate of development and stage of the disease. In most cases, the clinical signs are those of the initiating cause. In horses, depression, colic, and diarrhea are common with oliguria or polyuria. Clinical

signs in cattle with uremia are similar and in addition are frequently recumbent, and in severe and terminal cases cattle may have a bleeding diathesis. In chronic renal disease of all species, there is a severe loss of BW, weakness, anorexia, polyuria, polydipsia, and ventral edema.

UREMIA Uremia is the systemic state that occurs in the terminal stages of renal insufficiency. Anuria or oliguria may occur with uremia. Oliguria is more common unless there is complete obstruction of the urinary tract. Chronic renal disease is usually manifested by polyuria, but oliguria appears in the terminal stages when clinical uremia devel­ ops. The uremic animal is depressed and anorexic with muscular weakness and tremor. In chronic uremia, the body con­ dition is poor, probably as a result of contin­ ued loss of protein in the urine, dehydration, and anorexia. The respiration is usually increased in rate and depth but is not dys­ pneic; in the terminal stages it may become periodic in character. The heart rate is mark­ edly increased because of terminal dehydra­ tion, but the rectal temperature remains normal except in infectious processes and some cases of acute tubular nephrosis. An ammoniacal or uriniferous smell on the breath is often described in textbooks but is rarely clinically detectable. Uremic encephalopathy occurs in a small proportion of cattle, goats, and horses with chronic renal failure that involves an unknown metabolic pathway. It is associated with seizures, tremors, abnormal behavior, and muscle weakness, and histologic evidence of myelin vacuolation may be present.1 The animal becomes recumbent and comatose in the terminal stages of uremia. The temperature falls to below normal and death occurs quietly; the whole course of the disease is one of gradual intoxication. Necropsy findings, apart from those of the primary disease, are nonspecific and include degeneration of parenchymatous organs, sometimes accompanied by emaciation and moderate gastroenteritis. Uremia has been produced experimen­ tally in cattle by bilateral nephrectomy and urethral ligation. There is a progressive increase in serum urea concentration (mean daily increase of 53 mg/dL), serum creati­ nine concentration (mean daily increase of approximately 3.5 mg/dL), and serum uric acid concentration. Similar findings are reported in prerenal uremia in cattle. Inter­ estingly, serum phosphate and potassium concentrations were for the most part unchanged because of increased salivary secretion of both factors, and acidemia and metabolic acidosis were not evident. Serum potassium concentrations were mildly increased after 5 to 7 days of bilateral nephrectomy.

Special Examination of the Urinary System

VetBooks.ir

Special Examination of the Urinary System Lack of accessibility limits the value of physi­ cal examination of the urinary tract in farm animals. Palpation per rectum can be per­ formed on horses and cattle and is described in Chapter’s 7 and 8. In small ruminants and calves the urinary system is largely inacces­ sible to physical examination, although the kidneys may be palpated transabdominally and the urethra palpated digitally with the finger for periodic contractions that are common in male sheep and goats with obstructive urolithiasis. Urinalysis and determination of the serum or plasma con­ centration of urea nitrogen or creatinine is a required component of any examination of the urinary system.

TESTS OF RENAL FUNCTION AND DETECTION OF RENAL INJURY The simplest and most important test of urinary function is the determination of whether or not urine is being voided. This can be accomplished in large animals by keeping them on a clean, dry floor that is examined periodically. Placing an absorbent cloth under recumbent foals and calves will also help determine whether urine is being passed. Renal function tests evaluate the func­ tional capability of the kidney and generally assess blood flow to the kidneys, glomerular filtration, and tubular function. These tests depend on whether they are based on the examination of serum/plasma, urine, or both, and assess either function or the pres­ ence of injury. The most practical screening tests for the presence of decreased renal function are determination of serum creati­ nine concentration and urine specific gravity. Determination of both factors assists differ­ entiation of renal azotemia from prerenal azotemia. In prerenal azotemia, tubular function remains intact and renal conserva­ tion of water is optimized, resulting in the production of concentrated urine. Animals with prerenal azotemia therefore have increased serum concentrations of creatinine and urea and increased urine specific gravity. For comparison, animals with some degree of renal azotemia have increased serum con­ centrations of creatinine and urea and a lower than expected value for urine specific gravity. Determination of urine specific gravity should therefore be routinely per­ formed in all dehydrated animals before the initiation of treatment, because oral or intra­ venous (IV) fluid therapy will directly change urine specific gravity.

COLLECTION OF URINE SAMPLES Collection of urine samples can be difficult. Free-flow and catheterized samples are

equally useful for routine urinalysis. Urine samples for analysis should be collected by midstream voiding, or cystocentesis in small male ruminants, preferably with ultrasono­ graphic guidance. Bethanechol (0.075 mg/kg subcutaneously) has occasionally been used to produce urine in reluctant individuals, but a spontaneously voided sample is preferred for initial screening, which is routinely done using urine dipsticks. Horses will often urinate shortly after they are walked into a freshly bedded box stall. Cows urinate if they are relaxed and have their perineum and vulval tip massaged upward very gently, without touching the tail. Success rates in obtaining a urine sample can approach 100% if cows are recumbent and quietly encouraged to stand before attempting perineal stimulation to induce urination. Steers and bulls may urinate if the preputial orifice is massaged and splashed with warm water. Ewes often urinate imme­ diately after rising if they have been recum­ bent for some time. Occluding their nostrils and threatening asphyxia may also induce urination just as they are released and allowed to breathe again; however, this is a stressful procedure and should not be per­ formed in sick or debilitated sheep. An IV injection of furosemide (0.5–1.0 mg/kg BW) produces urination in most animals in about 20 minutes. The sample is useful for micro­ biologic examination but its composition has been drastically altered by the diuretic. Diuretics should be used with extreme caution in dehydrated animals. Urine samples obtained by bladder catheterization using a urethral catheter are

1099

preferred for microbiologic examination, provided aseptic technique is applied, includ­ ing bandaging the tail of female horses or holding the tail of cattle out of the way. The perineal region should then be cleaned with dilute povidone iodine or chlorhexidine to minimize urinary tract contamination, and there should be routine use of sterile surgical gloves and lubrication. Rams, boars, and young calves usually cannot be catheterized without fluoroscopy because of the presence of a suburethral diverticulum and the small diameter of the urethra. A precurved cathe­ ter and fluoroscopic guidance can be used to facilitate catheterization of rams and bucks. Ewes and sows can be catheterized, but their vulvas are often too small relative to hand size to allow access to the urethra. Cows can be catheterized relatively simply provided that a fairly rigid, small-diameter (0.5-cm) catheter is used, such as an artificial insemi­ nation pipette. A finger can be inserted into the suburethral diverticulum on the ventral aspect to direct the tip of the catheter over the diverticulum and into the external ure­ thral orifice (Fig. 13-1). For longer term catheterization of adult cattle (3 days), 24- to 28-French Foley catheters are placed into the bladder using the same insertion method; however, insertion of Foley catheters is facili­ tated by application of sterile lube on the outside of the catheter and placement of an insemination pipette into the catheter lumen to increase rigidity. Retention of Foley cath­ eters in cows is facilitated by using a balloon volume of 60 to 75 mL; use of smaller balloon volumes permits the catheter to move into the urethra, leading to pollakiuria,

b

a

c

Fig. 13-1  Lateral view of the urethra (a), vagina (b), and suburethral diverticulum (c) in adult female cattle as viewed from the left. A finger is inserted into the suburethral diverticulum on the ventral aspect to direct the tip of the catheter over the diverticulum and into the external urethral orifice. (Reprinted with permission from Rosenberger G. Clinical examination of cattle. Berlin: Parey; 1979; 453.)

1100

Chapter 13 

■ 

Diseases of the Urinary System

VetBooks.ir

stranguria, and potential catheter extrusion. The incidence of urinary tract infection with indwelling Foley catheters in dairy cows is 3% per catheterized day;2 the urine should therefore be periodically examined for evi­ dence of cystitis and antimicrobial treatment instituted whenever indicated. Mares can be catheterized easily, either by blindly passing a rigid catheter into the external urethral orifice or by using a finger as a guide for a flexible catheter. Long-term catheterization of the bladder of the mare requires a similar technique to that described for cows. Male horses can also be catheterized easily if the penis is relaxed. When urethral obstruction is present, the penis is usually relaxed, but administration of an ataractic drug (acepromazine is often used) makes manipulation of the penis easier and often results in its complete relaxation. Because of the long urethra, the catheter must be well lubricated. The catheter should be rigid enough to pass through the long urethra but flexible enough to pass around the ischial arch. In all species, catheterization over­ comes the natural defense mechanisms that prevent infectious organisms from ascend­ ing the urinary tract. As a result, attention to hygiene during catheterization is essential.

TESTS OF URINE SAMPLES Urinalysis is an essential component of the examination of the urinary system. The reader is referred to a textbook of veterinary clinical pathology for details of the biochem­ ical and microscopic examination of the urine. Cytologic examination of urine should take place as soon after collection as possible because casts (cylindrically shaped molds that indicate tubular injury) are fragile and can rapidly disintegrate. The common abnor­ malities of urine are discussed later. The urine sample should be centrifuged; the supernatant should be used for labora­ tory analysis and the sediment and remain­ ing supernatant for routine urine analysis. Specific Gravity Specific gravity of urine is the simplest test to measure the capacity of renal tubules to conserve fluid and excrete solute. For most species, the normal specific gravity range is 1.015 to 1.035, and in azotemic animals, spe­ cific gravity should be greater than 1.020 if the azotemia is prerenal in origin. In chronic renal disease the urine specific gravity decreases to 1.008 to 1.012 and is not appre­ ciably altered by either deprivation of water for 24 hours or the administration of large quantities of water by stomach tube. It is important to recognize that a specific gravity of less than 1.008 indicates that the kidney can produce dilute urine and, if sustained, indicates better renal function than a fixed urine specific gravity of 1.008 to 1.012. Specific gravity can be inaccurate when other refractive particles are present in urine,

such as glucose or protein. Urine specific gravity should therefore be used with caution in animals with proteinuria or glucosuria. As an alternative to specific gravity, osmolality of a fluid directly measures the concentration of solute in the fluid. Urine osmolality there­ fore provides a more accurate assessment of the tubule’s ability to conserve or excrete solute than urine specific gravity and is the preferred test of urine concentrating ability for research studies. However, urine specific gravity is sufficiently accurate for clinical use in animals without proteinuria or glucosuria, because there is a linear relationship between urine specific gravity and osmolality and urine specific gravity explains 52% of the variation in urine osmolality. The 95% con­ fidence interval for predicting osmolality from the specific gravity measurement is ±157 mOsmol/kg. pH The pH of urine can be measured using pH papers calibrated in 0.2 to 0.3 pH units or urine dipstick point-of-care tests that are calibrated in 0.5 pH units. The physiologic range of urine pH is 4.5 to 9.0, with herbivore urine typically being between 7.0 and 8.5. Cattle on high-grain diets may have slight aciduria (pH 6.0–7.0), and ruminants and horses ingesting an acidogenic diet will have aciduria, with urine pH as low as 5.0. Urine pH on free-catch samples is typically 0.1 to 0.2 pH units lower than anaerobically col­ lected samples; the difference is most likely caused by the loss of CO2 from urine during voided, which is accompanied by an increase in pH. It is for this reason that some research studies collect urine using a Foley catheter into a glass jar with mineral oil on the surface. For clinical use, it is sufficient to completely fill a screw top container with urine and minimize the air at the top of the container before urine pH is measured. An interesting and consistent finding is that aciduria is always accompanied by increased urine excretion of calcium. Low luminal pH in the distal convoluted tubule and connecting tubule decreases the number of epithelial Ca channels (transient receptor potential vanilloid member 5 [TRPV5]); the TRPV5 channel is considered to be the primary gatekeeper of active calcium reab­ sorption in the distal region of the urinary tract. Low luminal pH also decreases the pore size of the TRPV5 channel, resulting in decreased calcium uptake from the tubular lumen into the epithelial cell. The low luminal pH-induced decrease in TRPV5 number and activity result in decreased calcium absorption in the distal convoluted tubule and connecting tubule, directly result­ ing in hypercalciuria. Net Acid Excretion The kidney plays a central role in acid-base homeostasis by adjusting urine electrolyte excretion to maintain constant blood pH.

Measurement of urinary net acid excretion (NAE) provides a sensitive and clinically useful method for evaluating acid-base balance. This is because NAE provides an estimate of endogenous acid production and the magnitude of dietary acidification when an acidogenic diet is fed. The term NAE is commonly used in studies of renal physiol­ ogy in humans, other omnivores, and carni­ vores, in which urine pH is typically acidic, compared with plasma pH in a healthy animal (7.40). The term net base excretion (NBE) is more appropriate in cattle and other herbivores because urine pH is usually alka­ line. It should be recognized that NBE = −NAE, with both measured in milliequiva­ lents per liter.3 Urinary NAE is the most sensitive index of acid-base status and is clinically underuti­ lized. The Jørgensen method is often used to measure NAE and involves laboratory titra­ tion of urine to a standardized endpoint in which the temperature is 37°C, Pco2 is 0 mm Hg, and pH is 7.40 (equivalent to plasma pH in a clinically normal animal). The method involves acidification of the urine sample to pH 5 years) and immune status (there are no reports of horses affected twice by the disease, suggesting long-lasting immunity). • Introduction of EHV-1: almost always associated with a horse shedding the virus, either as a result of new infection or recrudescence of latent infection. • Presence of the D752 variant: although disease can occur associated with infection by N752. • Season: there appears to be higher incidences of the disease in the Northern Hemisphere in autumn, winter, and spring. • Pyrexia: horses that are pyrexic during an outbreak are more likely to develop EHM. • Movement of new horses onto the property, or use of horses in riding schools.32 • Possible associations with sex (increased risk if female) or breed (pony), although these associations are not consistent in all or most studies and are of limited usefulness in controlling or managing the disease.43,46,47

1276

Chapter 14 

■ 

Diseases of the Nervous System

VetBooks.ir

Immunity Immunity to EHV-1 is mediated by cytotoxic T cells, which explains the limited efficacy of inactivated virus vaccines that have minimal effect in stimulating cytotoxic T cells despite being capable of inducing a humoral immune response.52 The presence of EHV-1 cytotoxic T-cell precursors correlates well with protection from experimental infection, and some of the EHV-1 antigens responsible for this resistance have been identified.53-55 Mares usually only abort from EHV-1 infection once in their lifetime, and there are no reports of horses developing myeloencephalopathy more than once. Lack of antibodies to EHV-1 was identified as a risk factor in an outbreak of EHM in a herd of mares with foals at foot. Mares with strong antibody responses to EHV-1 did not develop disease. Economic Importance Disease associated with EHV-1 is of considerable economic importance because of the loss of training time and opportunities to perform during convalescence and quarantine, the loss of pregnancies during abortion storms, and deaths caused by myeloencephalopathy and infection of neonates.

PATHOGENESIS

The three organ systems involved in clinical disease associated with EHV-1 infection are the respiratory tract, uterus and placenta, and CNS. The common final pathway for injury in each of these body systems is damage to vascular endothelium with subsequent necrosis, thrombosis, and ischemia. Following EHV-1 exposure to the upper respiratory tract, virus can be detected in the soft palate and mainstem bronchus within 12 hours, and at all levels of the respiratory tract by 24 hours. The virus gains access to the body after binding to respiratory mucosal epithelium where it forms plaques that do not extend into submucosal tissues.35 In the respiratory tract there is an initial phase after infection of nasal epithelium56 in which there is rapid proliferation of the virus in the nasal, pharyngeal, and tonsillar mucosae, with subsequent penetration and infection of local blood vessels. This is followed by a systemic, viremic phase in which the virus is closely associated with blood lymphocytes (especially CD172a(+)),56 from which it can be isolated. Infection induces increased production of IFN-γ by T lymphocytes.54 Absence of viral antigens on the surface of EHV-1– infected peripheral blood mononuclear cells explains their ability to avoid complementmediated lysis. This activity, combined with the immunosuppression that accompanies EHV-1 infection,55,57-59 allows dissemination of the infection to the reproductive tract and CNS. Immunosuppression is mediated by production in EHV-1–infected cells of an “early protein” that interferes with peptide

translocation by the transporter associated with antigen processing. Immunosuppression is evident as reduced in vitro proliferation of peripheral blood monocytes and downregulation of expression of major histocompatibility complex class I molecules on the surface of infected cells. It is from this point that the invasion of lungs, placenta, fetus, and nervous tissue occur. Movement of infected mononuclear cells into target tissues is associated with expression of adhesion molecules by endothelium in the gravid uterus and in leukocytes. Viral infection of endothelium results in death of endothelial cells, inflammation, activation of clotting factors and platelets, increases in markers of fibrin degradation, and formation of blood clots in small vessels.60-62 This thrombotic disease causes ischemia of neighboring tissues with subsequent necrosis and loss of function. Another theory is that deposition of antigen–antibody complexes in small vessels results in an Arthus reaction with subsequent ischemia, necrosis, and loss of function. However, recent demonstration that mares with no antibody titer to EHV-1 were at increased risk of developing myeloencephalopathy does not support a role for type III hypersensitivity in this disease. Regardless of the underlying mechanism, clinical signs are a result of vasculitis and necrosis of tissue in the CNS and reproductive tract. This is in contrast to neurologic disease associated with herpesvirus in other species, in which the nervous system disease is a direct result of infection of neural tissues. Abortion is caused by damage to the placenta, endometrium, or fetus. Placental lesions include vasculitis, focal thrombosis, and infarction of the microcotyledons of the pregnant uterus. The fetus is infected and there are diagnostic lesions present in many aborted foals, including massive destruction of lymphocytes in the spleen and the thymus. In those abortions in which there is no lesion or evidence of virus infection in the foal, there may be extensive damage to the endometrium caused by an endothelial lesion and its attendant vasculitis, thrombosis, and secondary ischemia. Foals that are infected in utero but survive to full term may be stillborn or weak and die soon after birth with pulmonary, hepatic, and cardiac lesions. EHV-1 infection in foals not infected before or at birth is usually a self-limiting, mild infection of the upper respiratory tract with an accompanying leukopenia and a transitory immune suppression, although uveitis and occasionally death occur in a small number of foals. Virus can be isolated from the nasal mucus and the buffy coat of the blood for some time after clinical signs have disappeared. The pathogenesis of myeloencephalopathy in horses contrasts with herpesvirus encephalitis of other species in which there is viral infection of neuronal tissue. The

myeloencephalopathy in horses is, as discussed earlier, the result of vasculitis, thrombosis, and subsequent ischemia of neural tissue. Impairment of blood flow results in hypoxia and dysfunction or death of adjacent neural tissue.

CLINICAL FINDINGS

EHV-1 infection manifests as several forms of disease on a farm such that nervous system involvement can occur in an outbreak in which abortion and respiratory disease also feature, although more commonly one form of the disease (myeloencephalopathy or abortion) occurs alone or with mild respiratory disease. Foals, stallions, and mares can be affected with one or the other form of the disease, although it is most commonly seen in adult horses. Onset of neurologic signs is usually, but not invariably, preceded by cases of respiratory disease, fever, limb edema, or abortion. Myeloencephalopathy Myeloencephalopathy initially occurs in an index case, which might or might not have had signs of infectious respiratory disease alone or with signs of neurologic disease. Signs of neurologic disease develop in other horses approximately 6 to 14 days after disease in the index case. Disease then develops in a number of horses over a short period of time (3–10 days). Outbreaks in a stable can evolve rapidly.25,43,46,47 Fever, without signs of respiratory disease, often precedes signs of neurologic disease by 24 to 72 hours. The onset of neurologic signs is usually rapid, with the signs stabilizing within 1 to 2 days. Fever is more common (odds ratio 20×, 95% CI 3.4–390) in horses that go on to develop EHM, but the presence of limb edema or severity of nasal discharge are not associated with the likelihood of developing EHM during an outbreak of the disease.32,46 Thirteen percent of 61 horses with fever recorded during an outbreak of abortion and EHM developed signs of EHM.25 Six of seven pregnant mares aborted. Signs are variable but usually referable to spinal white matter involvement. Affected horses have variable degrees of ataxia and paresis manifest as stumbling, toe dragging, pivoting, and circumduction that is most severe in the hindlimbs. Signs are usually symmetric. There is often hypotonia of the tail and anus. Fecal and urinary incontinence are common and affected horses often dribble urine, have urine scalding of the skin of the perineum and legs, and require manual evacuation of the rectum. The severity of signs can progress to hemiplegia or paraplegia manifesting as recumbency and the inability to rise. Less commonly, CN deficits, such as lingual or pharyngeal paresis, head tilt, nystagmus, or strabismus, are present. Affected horses are usually alert and maintain their appetite.

Viral Diseases Primarily Affecting the Cerebrum

VetBooks.ir

Severity of neurologic disease varies among horses within an outbreak, and the prognosis is related to the severity of disease. In general, horses that become recumbent have a poor prognosis for both short-term and long-term survival despite intensive nursing care.43,46,47 However, less severely affected horses have a good prognosis for survival, with case–fatality rates as low as 2% to 3% in some outbreaks. Horses with mild signs of neurologic disease often recover completely and return to their previous level of performance, although some have persistent neurologic deficits after 1 year. Abortion Outbreaks of abortion might not be preceded by clinically apparent respiratory disease. The incidence of abortion is highest in the last third of pregnancy, particularly in the 8- to 10-month period but can occur as early as the fifth month. Abortion occurs without premonitory signs, and the placenta is usually not retained. Frequently there is no mammary development. Affected mares sometimes have prolapse of the uterus. Some foals are stillborn, whereas others are weak and die soon after birth. Abortion storms are often long-lasting, with a period of 17 to 22 days separating the index case from cases caused by secondary transmission of the virus, suggesting an incubation period of 2 to 3 weeks. Experimental infections induce abortion 15 to 65

days after intranasal inoculation of the virus. Although most abortions then occur within 1 month of the first secondary cases, abortions on a farm can continue for many months.27 Neonatal Viremia and Septicemia In utero EHV-1 infection causes abortion or the birth of infected foals, some of which are normal at birth, but become weak and die within 3 to 7 days of birth with signs of respiratory distress and septicemia. A less severe form of the disease, characterized by pyrexia, nasal discharge, and chorioretinitis, occurs in slightly older foals that are apparently infected after birth. Affected foals that survive sometimes do not have serum antibodies to EHV-1. Death may be associated with secondary bacterial infection with E. coli or Actinobacillus equuli, although EHV-1 infection alone is sufficient to cause death. Respiratory Disease The classical respiratory tract form of the disease (rhinopneumonitis) is virtually indistinguishable on the basis of clinical signs from the other upper respiratory tract diseases of horses and is identical to that associated with EHV-4.

CLINICAL PATHOLOGY

Results of hematologic and serum biochemical examinations are neither specific nor diagnostic. EHV-1 infection of adult horses

1277

results in leukopenia that is attributable to both neutropenia and T-cell lymphopenia, with B-cell lymphocytosis occurring during the recovery period. EHV-1 septicemia of foals is characterized by profound leukopenia, neutropenia with a left shift, and lymphopenia. An approach to achieving prompt antemortem diagnosis of EHM is suggested in Fig. 14-9.63 CSF of horses with EHV-1 encephalomyelopathy is characteristically xanthochromic and has an increased total protein concentration (>1 g/L) with a normal white cell count.32,64 The interpretation of EHV-1 antibody in CSF is uncertain, although normal horses are not expected to have detectable antibodies to EHV-1 in the CSF. Serologic tests are of critical importance in diagnosis and control of EHV infections. Many horses have serum antibodies to EHV-1 and EHV-4 as a result of previous infection or vaccination. Thus the demonstration of antibodies is not in itself sufficient to confirm a diagnosis of the disease. Complement-fixing antibody appears on the 10th to 12th day after experimental infection but persists for only a limited period. Demonstration of a threefold to fourfold increase in the serum concentration of specific complement-fixing antibodies in acute and convalescent serum samples provides persuasive evidence of recent infection. Complement-fixing antibodies persist for only a short time (several months) while VN

Fig. 14-9  Methodology for rapid antemortem diagnosis of equine herpesvirus-1 (EHV-1) myeloencephalopathy in horses with signs of nervous system disease. Solid lines represent a diagnostic pathway. EDTA, ethylenediaminetetraacetic acid. (Reproduced, with permission, from Pusterla N, Wilson WD, Madigan JE, Ferraro GL. Equine herpesvirus-1 myeloencephalopathy: a review of recent developments. Vet J 2009;180:279-289.)

1278

Chapter 14 

■ 

Diseases of the Nervous System

VetBooks.ir

antibodies persist for over a year, and testing for them is therefore a more reliable means of determining that previous infection with the virus has occurred. Until recently, serologic differentiation of antibodies to EHV-1 and EHV-4 was not possible. However, highly specific ELISA tests based on differences between EHV-1 and EHV-2 in the variable region of the C terminus of glycoprotein G, at least one of which is commercially available, have been developed that can differentiate between antibodies to EHV-1 and EHV-4 in horse serum. The ELISA is reported to be more sensitive, easier to perform, more rapid, and more reproducible than the virus neutralization test. Importantly, the ELISA test is able to differentiate between infections associated with EHV-1 and EHV-4.65,66 Identification of the virus in nasal swabs, or blood buffy coat, or tissue by culture or a PCR test provides confirmation of infection.67-71 The use of seminested or multiplex PCR or qPCR, which avoids the risk of carryover contamination, provides rapid identification of EHV-1 viral genome in nasopharyngeal swabs, blood, and other tissues. The test is at least as sensitive as viral isolation in identifying presence of virus. Rapid identification of virus shedding using qPCR can facilitate monitoring and interventions to prevent spread of infection and additional examination or prophylactic treatment of infected horses. Appropriate PCR testing can determine whether the EHV-1 is the D752 or N752 variant. This information can be important in epidemiologic investigations and might have implications for administration of antiviral therapy, although this is unclear, but generally does not influence management of a disease outbreak.21,72 The virus can be isolated in tissue culture, chick embryos and hamsters, from either nasal washings or aborted fetuses, and has growth characteristics that differentiate it from EHV-4.73 Samples of nasopharyngeal exudate for virus isolation are best obtained from horses during the very early, febrile stages of disease, and are collected via the nares by swabbing the nasopharyngeal area with a 5 × 5-cm gauze sponge attached to the end of a 50-cm length of flexible, stainless steel wire encased in latex rubber tubing. A guarded uterine swab devise can also be used. After collection, the swab should be removed from the wire and transported promptly to the virology laboratory in 3 mL of cold (not frozen) fluid transport medium (serum-free minimal essential medium with antibiotics). Virus infectivity can be prolonged by the addition of bovine serum albumin or gelatin to 0.1% (w/v).

NECROPSY FINDINGS

Macroscopic findings in aborted fetuses include petechial and ecchymotic hem

orrhages, especially beneath the respiratory mucosae. The most consistent finding is an excess of clear yellow fluid in the pleural and peritoneal cavities. Focal hepatic necrosis and slight icterus may also be present. In some aborted fetuses the cut surface of the spleen reveals unusually prominent lymphoid follicles, which are swollen from necrosis and edema. Acidophilic intranuclear inclusion bodies may be evident histologically in a variety of cell types, including the bronchiolar and alveolar epithelium, hepatocytes, and dendritic cells of the lymphoid tissues. Although the microscopic pathology is unimpressive, examination of the placenta via IHC techniques can be a useful aid in the diagnosis of EHV-1–induced and EHV-4–induced abortions. In foals that are alive at birth but die soon afterward there is usually massive pulmonary congestion and edema, with collapse of the lung and hyaline membrane development in those that survive longer. In the nervous or paralytic form of the disease there is an acute disseminated myeloencephalopathy. Hemorrhages may be visible grossly but often there are no macroscopic changes. Disseminated vasculitis occurs in the experimental disease, and the malacic lesions present in the nervous tissue are the result of leakage from these damaged vessels. The virus can be isolated from the brain, and the isolation is facilitated by use of an indirect peroxidase stain to establish the location of the virus. The virus infects endothelial cells within the CNS but has also been demonstrated within neurons and astrocytes and has been linked to chorioretinitis in a foal. In rare cases the virus may cause lesions in other tissues, such as the intestinal mucosa and spleen or pharynx. The laboratory examination of aborted fetuses should include a search for virus by tissue culture and IHC or PCR techniques, as well as a histologic examination of the lung and liver for the presence of inclusion bodies. A direct FAT has also been used. A serologic examination of the foal may provide useful information in those cases in which attempts at isolation are negative but seroconversion has occurred. However, a recent study found that fetal serology was an unreliable means of diagnosing EHV-1 abortion, and that IHC was slightly more sensitive than virus isolation. Samples for Confirmation of Diagnosis • Virology: chilled lung, liver, spleen, thymus, and thoracic fluid of aborted fetuses or neonates. Spinal cord or brain of horses with nervous disease (VI, PCR, FAT, serology). • Histology: fixed lung, liver, spleen, thymus, and trachea from fetuses or neonates. • Fixed brain and spinal cord from several sites, as well as Bouin’s fixed eye should

be examined in adults with nervous disease (LM, IHC). DIFFERENTIAL DIAGNOSIS Respiratory disease in horses is associated with a variety of agents (Table 12-14). Abortion can be associated with leptospirosis, Salmonella abortusequi, placentitis associated with Streptococcus zooepidemicus or Escherichia coli, associated with mare reproductive loss syndrome, or congenital abnormalities, among other causes. When other pregnant mares are at risk, abortion in a late-term mare should always be considered to be caused by EHV-1 until proved otherwise. Neurologic diseases with clinical presentations similar to that associated with EHV-1 include rabies, equine protozoal myeloencephalitis, neuritis of the cauda equina (equine polyneuritis), trauma, acute spinal cord compression (cervical stenotic myelopathy), and equine degenerative myelopathy. Fever is rare in other neurologic diseases of horses, and any horse with neurologic disease and fever or a history of fever within the previous week should be considered to have EHV-1 myeloencephalopathy. Outbreaks of posterior paresis or ataxia, especially in horses without fever, should prompt consideration of ingestion of intoxicants such as Astragalus spp., Swainsona spp., or sorghum. Ryegrass staggers can produce similar signs of ataxia. Neonatal septicemia can be associated with E. coli, Streptococci spp., and other bacteria, especially in foals with failure of transfer of maternal immunoglobulins. EHV-1, equid herphesvirus-1.

TREATMENT Because of the highly contagious nature of EHV-1 infections, horses with respiratory disease, abortion, or neurologic disease, especially if these occur as an outbreak, should be isolated until the cause of the disease is identified. There is no specific treatment for the diseases associated with EHV infection, although acyclovir and other antiviral drugs are used on occasion to treat horses in outbreaks of myeloencephalopathy.46 Horses with EHM require intense supportive care. Nursing care to prevent urine scalding, pressure sores, and pneumonia is important in horses with myeloencephalopathy. Recumbent or severely ataxic horses should be supported to stand if at all possible. Although a rope tied to the tail and slung over an overhead beam may be used to assist the horse to stand, a sling may be necessary to support more severely affected horses. Nursing care is important to prevent development of pressure sores in recumbent horses or those supported by slings. The perineum of incontinent horses should be cleaned frequently, and salves or ointments

Viral Diseases Primarily Affecting the Cerebrum

VetBooks.ir

to protect the skin applied. Some horses require catheterization of the bladder to relieve distension. Enemas, accompanied by careful manual evacuation of the rectum, might be needed to promote passage of feces. Administration of corticosteroids to these horses is controversial, but many clinicians administer dexamethasone sodium phosphate (0.05–0.25 mg/kg intramuscularly every 12–24 hours) or prednisolone (1–2 mg/kg orally or parenterally every 24 hours) for 2 to 3 days. Administration of corticosteroids may be contraindicated because of the presence of replicating virus in affected horses. The use of antiplatelet drugs or antithrombotic compounds has received anecdotal support, but there is no evidence that they do not harm affected horses and similarly no evidence of efficacy. Administration of drugs to inhibit viral replication has merit and is attempted during outbreaks of disease. The challenges of this approach are that the infection is well advanced by the time clinical signs of neurologic disease are detected, especially in cases early in the disease outbreak before purposeful monitoring is in place, pharmacokinetics and pharmacodynamics of the available drugs are unknown or imperfectly known, and the drugs are expensive. Antiviral drugs considered for use in horses with EHM include acyclovir, valacyclovir, penciclovir (after oral administration of its prodrug famciclovir), ganciclovir, and valganciclovir.74-78 Acyclovir is effective against EHV-1 in vitro, and pharmacokinetic studies suggest that administration of 10 mg/kg orally every 4 to 6 hours (five times daily) or 10 mg/kg intravenously every 8 hours results in acceptable concentrations of drug in the blood. However, further investigation reveals that there is a large variation between individual horses in the absorption of acyclovir with consequent failure to obtain therapeutic concentrations in many horses.79 The in vitro activity of acyclovir, ganciclovir, cidofovir, adefovir, 9-(2-phosphonylmethoxyethyl)2,6-diaminopurine (PMEDAP) and foscarnet against three abortigenic isolates and three neuropathogenic isolates of EHV-1 revealed variable activity of cidofovir and limited to no activity of foscarnet.80 Current recommendations for the prophylaxis and treatment of horses with EHM include administration of acyclovir (10–20 mg/kg every 5–8 hours, orally for 7 days) or ganciclovir IV at 2.5 mg/kg every 8 h for 24 h followed by maintenance dosing of 2.5 mg/kg every 12 h, or orally at 30–40 mg/kg every 8–12 h for 7 days.72 The efficacy of these compounds has not been demonstrated in appropriate clinical trials, and earlier comments about the variability in oral bioavailability of acyclovir should be noted. Neonatal foals with septicemia should be treated aggressively with antibiotics and supportive care, including enteral or

parenteral nutrition and fluid administration (see the section Clinical Assessment and Care of Critically Ill Newborns in Chapter 19). Treatment with acyclovir has been reported. Failure of transfer of passive immunity should be rectified with oral or intravenous administration of colostrum or plasma, respectively.

CONTROL

Recommendations for programs to prevent introduction of infection and to control EHM and abortion outbreaks are available from several sources and might vary between countries.18,21,29,81 Prevention of Infection The general principles include the following: • Enhanced immunity, currently attempted by vaccination • Subdivision and maintenance of the farm population in groups of horses to minimize spread of the infection • Minimize risk of introduction of infection by new horses • Minimize risk of reactivation of latent infection in resident horses • Develop plans for implementation of these routine control measures, and for actions in the event of an abortion • Educate management and staff as to the importance of strict adherence to these procedures The relative importance of each of these measures has not been determined, but implementation of control measures, including allocation of mares to small bands based on anticipated foaling date, quarantine of new introductions, and vaccination of pregnant mares, has reduced the incidence of EHV-1 abortion in central Kentucky. The most striking association has been an apparent reduction in the incidence of abortion storms. It must be emphasized that vaccination does not replace any of the other management procedures in control of this disease and that abortions have occurred among vaccinated mares on farms on which the other management procedures have been ignored. Vaccination Vaccination against respiratory disease and abortion associated with EHV-1 is widely practiced despite lack of clear-cut evidence that vaccination reduces the incidence or severity of either of these diseases. Information regarding field efficacy of EHV vaccines is lacking, and that derived from experimental challenge models is often contradictory or incomplete. Give these caveats, the following recommendations are made based on generally accepted practices. None of the currently available vaccines, of which there are approximately 14 worldwide, consistently prevent infection of vaccinated horses or provide complete protection against disease associated with EHV-1.21,52,72

1279

The principal objective of vaccination has been to protect mares against abortion associated with EHV-1, although vaccines intended to prevent rhinopneumonitis and containing both EHV-1 and EHV-4 are available. Additionally, vaccination of mares is intended to reduce transmission of EHV-1 to foals in an attempt to interrupt the cyclical nature of infection on stud farms. Vaccines consisting of a modified live EHV-1, inactivated EHV-1, or a mixture of inactivated EHV-1 and EHV-4 are available for intramuscular or intranasal administration to horses. Both inactivated and modified live EHV-1 vaccines elicit virus-neutralization and complement fixation antibody responses in horses, although high antibody titers are not necessarily related to resistance to infection. Resistance to infection might be more closely related to cytotoxic T-cell responses. Widespread use of a combined EHV-1 and EHV-4 killed virus vaccine in Australia has not reduced serologic evidence of infection in foals on farms where mares are vaccinated, although the vaccine was effective in preventing disease induced by experimental infection. Complicating assessment of vaccine efficacy is the variable response to vaccination by some mares and foals, with certain animals having minimal responses to vaccination, which in other horses elicits a strong immune response. Efforts are underway to develop modified live vaccines that can be administered intranasally. Intranasal administration of one such EHV-1 vaccine induced protection against experimentally induced EHV-1 (and EHV-4) respiratory disease and abortion in mares, and prevented infection of foals even when administered in the presence of maternally derived antibodies. An alternative approach is the development of subunit vaccines using the envelope glycoprotein D, which has been shown to elicit protective immunity in laboratory animal models of EHV-1 disease and administration of which induces VN antibody and glycoprotein D–specific ELISA antibodies in horses. Current modified live vaccines appear to induce a more restricted IgG isotype than does natural infection, which could partly account for their limited efficacy.53 Despite the incomplete protection afforded by vaccines, vaccination against EHV-1 is an important part of most equine herd health programs in the vaccination of pregnant and nonpregnant mares, foals, and adult horses. The intent of vaccination of mares is to prevent abortion associated with EHV-1. One inactivated virus vaccine is reported to decrease the incidence of abortion by 65%, although others have not been able to replicate this success and there are reports of abortion storms on farms of wellvaccinated mares. An inactivated virus vaccine containing EHV-1 and EHV-4 prevented abortion in five of six mares exposed experimentally to EHV-1, whereas all six nonvaccinated mares aborted. Mares are

1280

Chapter 14 

■ 

Diseases of the Nervous System

VetBooks.ir

vaccinated with the inactivated vaccine during the fifth, seventh, and ninth months of gestation. Additional vaccinations at breeding and 1 month before foaling are recommended by some authorities. No vaccines are currently licensed with the claim of preventing EHM, and the disease occurs in well-vaccinated horses. Concerns that the disease might represent a “second hit” as a result of vaccination and subsequent infection have not received widespread support and do not have empirical evidence that is in any way supportive.21 Foals are an important source of infection and control of infection in foals is considered critical to control of infection on a farm. Consequently, attention has been paid to the responses of foals to vaccination at various ages, given the risk of passive immunity interfering with vaccination and the early age at which foals are infected by EHV-1. Current recommendations vary with some authorities recommending vaccination of foals after 5 months of age, to avoid the interfering effect of passive immunity on response to vaccination. However, vaccination of foals at this age likely misses the period of time when foals are first infected by EHV-1 from their dam or other mares in the band. One recommendation is that foals should be vaccinated in their third month, with revaccination 1 month and 6 months later. Modified live virus vaccine is given to foals at 3 to 4 months of age, and nonpregnant mares and other horses are given two doses administered 3 months apart followed by revaccination every 9 months. Because of the short duration of immunity following vaccination, frequent vaccination, perhaps at intervals as short as 3 months, of horses at high risk is recommended. However, the efficacy of such a program is uncertain. Subdivision of Horses on a Farm Maintenance of small groups of horses of similar age and reproductive status is recommended to minimize the chances of spread of infection. Pregnant mares, after weaning of foals, should be maintained in a herd that does not have access to foals, weanlings, nonpregnant mares, or other equids (donkeys). Similarly, weaned foals should be separated from horses of other ages in recognition of the high rate of infection and viral shedding in weanlings. Failure to adhere to these procedures can result in rapid spread of infection and abortions among at-risk mares. Pregnant mares should be combined into small groups (~10) early in pregnancy based on their anticipated foaling dates. Multiparous mares should not be mixed with mares that are pregnant for the first time. Management practices should be introduced that minimize the opportunities for viral spread. Ideally, pregnant mares are handled using facilities separate from those used to handle mares with foals or weanlings. If common facilities must be used,

pregnant mares should be handled first, after thorough cleaning of the facility, followed by mares with foals and finally weanlings and other horses. Minimize Risk of Introduction   of Infection The only sources of virus are recrudescence of latent infection and introduction by newly arrived horses shedding virus. All horses must be considered as potentially shedding EHV-1 on arrival at a farm and should be isolated from resident horses. Introduction of new horses to the small groups of pregnant mares should be avoided if at all possible, or if absolutely necessary preceded by a 21-day isolation period. If at all possible, avoid mingling resident and nonresident mares even after quarantine of nonresident animals. Prevention of Reactivation of   Latent Infection The factors inciting reactivation of latent infection and viral shedding are unknown. However, stressful events, such as transportation or other disease, have the potential to cause reactivation of latent infection. For this reason pregnant mares should not be shipped within 8 weeks of expected foaling and all efforts, including vaccination, should be made to prevent other infectious diseases. Control of Outbreaks The principles underlying control of abortions or EHM caused by EHV-1 include the following: • Early and rapid diagnosis • Prevention of spread of infection • Treatment of individual cases These aims are approached through six stages: 1. Preliminary recognition of the problem (outbreak): typically by owners or trainers recognizing the presence of sick horses. 2. Preliminary veterinary investigation: conducted by a veterinarian on, usually, their first response to the owner’s concerns and leading to a presumptive clinical diagnosis. 3. Establishing the diagnosis: use of appropriate laboratory and other testing to confirm or rule out specific diagnoses. 4. Understanding and managing the outbreak: this is complex because it involves an understanding of the biology and epidemiology of the disease, the financial and social context of the outbreak, and assessment of the feasibility, and cost-effectiveness, of potential interventions. 5. Establishing freedom of infection: documenting the end of the outbreak and confirming freedom from infection by the offending agent.

6. Return the premise to normal function and activity. Control of Outbreaks of Myeloencephalopathy Diagnostic criteria for EHM are set out in the six stages list earlier. Adult horses with rapid onset of signs of nervous system disease, with or without fever, should be considered to have EHM until proven otherwise. Outbreaks of EHV-1–induced neurologic disease often occur in riding schools and similar situations where there is constant movement of horses on and off the property. As such it is exceedingly difficult to institute control measures that prevent introduction of the disease and that are compatible with the use of the horses. Having said that, the principles outlined earlier for preventing introduction of infection onto breeding farms also apply for prevention of myeloencephalopathy at riding stables. Reports of outbreaks of EHM in stables and veterinary hospitals have underscored the highly infectious nature of the disease.25,46,47 EHV-1 is spread from infected horses, which can have virus in nasal fluid before onset of clinical signs, by aerosol, and on fomites. It is critical to prevent spread by diligent attention to biosecurity, including spread by personnel and aerosol. Infected horses should be isolated in a separate air space to uninfected or at risk horses. Detailed instructions for handling outbreaks of neurologic disease attributable to EHV-1 are available and provide advice on quarantine, disinfection, and sample collection. There is no “one size fits all,” and the recommendations should be modified or adopted with a full understanding of the financial, social, and psychologic context of managing the outbreak. Guidelines for managing an outbreak of EHM include the following21,29,72,82: • Affected horses should be isolated because they are infectious. • The diagnosis should be confirmed by virus isolation, PCR, or histologic examination of tissues from affected horses that die or are euthanized. • Potentially affected horses should be tested to determine whether they are excreting the virus (nasal swabs). • There should be no movement of horses on or off the premises for at least 21 days after the last case has occurred. • Movement among bands of horses on the farm should be avoided. • Animals should leave or move between bands only when there is no evidence of continued active infection in their group. • Vaccination in the face of an outbreak of EHM is not recommended. Clinically affected horses should not be vaccinated. • Prophylactic use of acyclovir has been reported, although the efficacy of this practice is unknown.

Viral Diseases Primarily Affecting the Cerebrum

1281

VetBooks.ir

Table 14-13  Three-tiered approach to managing an outbreak of equine herpesvirus myeloencephalopathy. Three tiers of approach Action

Gold tier

Silver tier

Bronze tier

Segregate the population into small discrete groups that can be managed discretely to avoid infection transferring between them

Yes The smaller the groups the better to minimize the impact of ongoing disease and possibly reduce later laboratory test costs

Yes The smaller the groups the better to minimize the impact of ongoing disease and possibly reduce later laboratory test costs

Yes The smaller the groups the better to minimize the impact of ongoing disease and possibly reduce later laboratory test costs

Collect samples

Collect full set from all animals NP swab in VTM, serum (5–10 mL) and heparinized whole blood (30 ml)

Collect partial set from all animals NP swab in VTM and serum (5–10 mL)

Collect partial set from all animals NP swab in VTM and serum (5–10 mL)

Test samples

Test full set from all animals NP swab by qPCR, serum by CFT and heparinized blood by virus isolation

Test partial set from all animals NP swab by qPCR and serum by CFT

Do not test, but freeze the partial set from all animals for possible testing later

Observe for clinical disease (neurologic disease and/or abortion noting that pregnant mares should only be considered clear once they have a foaled successfully and have a healthy foal at foot)

Observe all groups for 3–4 weeks: If no clinical disease is observed in a group: collect NP swabs and sera (pair with already tested sample in CFT) and test, consider EHV-1 free if all results are negative If clinical disease is observed in a group: immediately collect and test a full set of samples from all horses in the affected group Remove positives to an isolation area Repeat after 2–3 weeks and only consider EHV-1 free when all results are negative

Observe all groups for 3–4 weeks: If no clinical disease is observed in a group: collect NP swabs and sera (pair with already tested sample in CFT) and test, consider EHV-1 free if all results are negative If clinical disease is observed in a group: immediately collect and test a full set of samples from all horses in the affected group Remove positives to an isolation area Repeat after 2–3 weeks and only consider EHV-1 free when all results are negative

Observe all groups for 3–4 weeks: If no clinical disease is observed in a group: collect NP swabs and sera (pair with frozen samples in CFT) and test, consider EHV-1 free if all results are negative If clinical disease is observed in a group: immediately collect a full set of samples from all the affected group and test all, including frozen, samples Remove positives to an isolation area Repeat after 2–3 weeks and only consider EHV-1 free when all results are negative

CFT, complement fixation test; NP, nasopharyngeal; qPCR, quantitative polymerase chain reaction; VTM, virus transport medium. Reproduced from Gonzalez-Medina S et al: Equine Vet J 2015; 47:142.

A suggested, three-tiered approach to managing an outbreak of EHM is depicted in Table 14-13. Abortion Rapid Diagnosis Every abortion in a late-term mare should be considered to be associated with EHV-1 until proven otherwise. Therefore rapid and early diagnosis of the abortion or of EHM is important to instituting control measures. In regions with large numbers of breeding mares, all abortions in mares should be investigated by detailed postmortem examination of the fetus and serologic examination of the mare. Prevention of Spread Diligent and concerted efforts must be made to prevent dissemination of infection from the initial focus in cases of abortion. Delay in doing so increases the incidence of abortion and prolongs the outbreak.27 Infected fetal tissues and fluids, and contaminated materials such as bedding, should be placed in impervious containers and either transported to a laboratory for examination or destroyed by incineration. Samples for laboratory examination should be handled to prevent spread of infection. Facilities and

equipment that might have been contaminated should be disinfected by thorough cleaning followed by application of a phenolic or iodophor disinfectant. The mare should be isolated until results of laboratory examination are negative for EHV-1 or until the second estrus, at which time it is unlikely that there is shedding of virus from the reproductive tract. Other mares in the same band as the mare that aborted should be considered exposed and at risk of abortion. These mares should be held in strict isolation until the results of laboratory examination are negative for EHV-1, or until they foal or abort. Other recommendations for horse movement include the following: • When an abortion occurs on the stud, no mares should be allowed to enter or leave it until the possibility of EHV-1 infection is excluded. However, maiden and barren mares, i.e., mares that have foaled normally at home but that are not in foal, coming from home studs where no signs of the disease are occurring, may be admitted because they are considered not to be infected. • If EHV-1 infection is identified on the stud, all pregnant mares ready to foal that season (i.e., late-pregnant mares)

should remain at the stud until they have foaled. The incubation period for EHV-1 abortion ranges between 9 and 121 days. • All nonpregnant animals and mares that have foaled should remain at the stud for 30 days after the last abortion. The main problem that arises in this program is in deciding what to do with mares that come into contact with the respiratory disease but not the abortion disease. This may occur very early in pregnancy and prolonged isolation would be onerous. The decision usually depends on the owner’s risk aversion and the availability of facilities to maintain long-term isolation. FURTHER READING Gonzalez-Medina S, Newton JR. Equine herpesvirus1:dealing pragmatically but effectively with an ever present threat. Equine Vet J. 2015;47:142-144. Lunn DP, et al. Equine herpesvirus-1 consensus statement. J Vet Intern Med. 2009;23:450-461. Pusterla N, Hussey GS. Equine herpesvirus 1 myeloencephalopathy. Vet Clin North Am Equine Pract. 2014;30:489-506.

REFERENCES

1. Davison AJ, et al. Arch Virol. 2009;154:171. 2. Schrenzel MD, et al. Emerg Infect Dis. 2008;14:1616. 3. Rebelo AR, et al. Can J Vet Res. 2015;79:155.

1282

Chapter 14 

■ 

Diseases of the Nervous System

VetBooks.ir

4. LeCuyer TE, et al. J Vet Diagn Invest. 2015;27:749. 5. De Witte FG, et al. J Vet Intern Med. 2012;26:1064. 6. Bell SA, et al. Vet Microbiol. 2008;130:176. 7. Rushton JO, et al. Vet J. 2014;200:200. 8. Vengust M, et al. J Vet Diagn Invest. 2008;20:820. 9. Pusterla N, et al. Vet Rec. 2010;167:376. 10. Wong D, et al. JAVMA. 2008;232:898. 11. Williams KJ, et al. PLoS ONE. 2013;8:e63535. 12. Vander Werf KA, et al. J Equine Vet Sci. 2014;34:738. 13. Vander Werf K, et al. J Vet Intern Med. 2013;27:387. 14. Hussey GS, et al. Vet Res. 2013;44:118. 15. Herder V, et al. Vet Microbiol. 2012;155:420. 16. Abdelgawad A, et al. PLoS ONE. 2015;10:e0138370. 17. Ibrahim ESM, et al. Arch Virol. 2007;152:245. 18. Dunowska M. New Zeal Vet J. 2014;62:171. 19. Ma G, et al. Vet Microbiol. 2013;167:123. 20. Nugent J, et al. J Virol. 2006;80:4047. 21. Lunn DP, et al. J Vet Intern Med. 2009;23:450. 22. Allen GP. Am J Vet Res. 2008;69:1595. 23. Pronost S, et al. Equine Vet J. 2010;42:672. 24. Pronost S, et al. Vet Microbiol. 2010;145:329. 25. Walter J, et al. Acta Vet Scand. 2013;55. 26. Stasiak K, et al. BMC Vet Res. 2015;11. 27. Schulman ML, et al. Equine Vet J. 2015;47:155. 28. Perkins GA, et al. Vet Microbiol. 2009;139:375. 29. Gonzalez-Medina S, et al. Equine Vet J. 2015;47:142. 30. Allen GP, et al. Equine Vet J. 2008;40:105. 31. Tsujimura K, et al. J Vet Med Sci. 2011;73:1663. 32. Burgess BA, et al. J Vet Intern Med. 2012;26:384. 33. Pusterla N, et al. Vet J. 2012;193:579. 34. Goodman LB, et al. PLoS Pathog. 2007;3:e160. 35. Vandekerckhove AP, et al. J Gen Virol. 2010;91:2019. 36. Estell KE, et al. Equine Vet J. 2015;47:689. 37. Wohlsein P, et al. Vet Microbiol. 2011;149:456. 38. Abdelgawad A, et al. Vet Microbiol. 2014;169:102. 39. Guo X, et al. J Vet Med Sci. 2014;76:1309. 40. Damiani AM, et al. Vet Microbiol. 2014;172:555. 41. Gryspeerdt A, et al. Vlaams Diergeneeskundig Tijdschr. 2011;80:147. 42. Mori E, et al. Rev - Off Int Epizoot. 2011;30:949. 43. van Galen G, et al. Vet Microbiol. 2015;179:304. 44. Bazanow BA, et al. Polish J Vet Sci. 2014;17:607. 45. Pronost S, et al. Transbound Emerg Dis. 2012;59:256. 46. Henninger RW, et al. J Vet Intern Med. 2007;21:157. 47. Goehring LS, et al. J Vet Intern Med. 2010;24:1176. 48. Gardiner DW, et al. Vaccine. 2012;30:6564. 49. Hebia I, et al. Theriogenology. 2007;67:1485. 50. Hebia-Fellah I, et al. Theriogenology. 2009;71:1381. 51. Pusterla N, et al. J Vet Intern Med. 2010;24:1153. 52. Paillot R, et al. Open Vet Sci J. 2008;2:68. 53. Goodman LB, et al. Clin Vaccine Immunol. 2012;19:235. 54. Paillot R, et al. Dev Comp Immunol. 2007;31:202. 55. Wimer CL, et al. Vet Immunol Immunopathol. 2011;140:266. 56. Gryspeerdt AC, et al. Vet Microbiol. 2010;142:242. 57. Luce R, et al. Equine Vet J. 2007;39:202. 58. Ma G, et al. J Virol. 2012;86:3554. 59. Sarkar S, et al. Vet Immunol Immunopathol. 2015;167:122. 60. Andoh K, et al. Virus Res. 2015;195:172. 61. Goehring LS, et al. J Vet Intern Med. 2013;27:1535. 62. Stokol T, et al. PLoS ONE. 2015;10:e0122640. 63. Pusterla N, et al. Vet J. 2009;180:279. 64. Goehring LS, et al. Vet J. 2010;186:180. 65. Amer HM, et al. Afr J Microbiol Res. 2011;5:4805. 66. Yildirim Y, et al. Iranian J Vet Res. 2015;16:341. 67. Hu Z, et al. Appl Microbiol Biotech. 2014;98:4179. 68. Pusterla N, et al. J Vet Diagn Invest. 2009;21:836. 69. Pusterla N, et al. Vet J. 2009;179:230. 70. Smith KL, et al. J Clin Microbiol. 2012;50:1981. 71. Stasiak K, et al. Polish J Vet Sci. 2015;18:833. 72. Pusterla N, et al. Vet Clin North Am Equine Pract. 2014;30:489. 73. Equine rhinopneumonitis (equine herpesvirus 1

and 4). OIE, 2015. (Accessed 07.02.2016, at http:// www.oie.int/fileadmin/Home/eng/Health_ standards/tahm/2.05.09_EQUINE_RHINO.pdf.). 74. Carmichael RJ, et al. J Vet Intern Med. 2010;24:712. 75. Carmichael RJ, et al. J Vet Pharmacol Ther. 2013;36:441. 76. Garre B, et al. Vet Microbiol. 2009;135:214. 77. Maxwell LK, et al. J Vet Pharmacol Ther. 2008;31:312. 78. Tsujimura K, et al. J Vet Med Sci. 2010;72:357. 79. Wong DM, et al. Equine Vet Educ. 2010;22:244. 80. Garre B, et al. Vet Microbiol. 2007;122:43. 81. Dunowska M. New Zeal Vet J. 2014;62:179. 82. Equine herpesvirus 1 and 4 related diseases. American Association of Equine Practitioners, 2013. (Accessed 07.02.2016, at http://www.aaep.org/ custdocs/EquineHerpesvirusFinal030513.pdf.).

PERUVIAN HORSE SICKNESS VIRUS Peruvian horse sickness virus is an Orbivirus associated with causing neurologic disease in horses in Peru with a mortality rate of approximately 1.25% and a case–fatality rate of 78%.1 A genetically identical virus has been isolated from horses dying of neurologic disease in northern Australia.2 Serologic surveillance in that area demonstrates antibody to Peruvian horse sickness virus in 11% of horses. The disease is described as causing motor incoordination, sagging jaw, tooth grinding, and stiff neck with death in 8 to 11 days. REFERENCES

1. Attoui H, et al. Virology. 2009;394:298. 2. Mendez-Lopez MR, et al. J Vector Ecol. 2015;40:355.

POWASSAN VIRUS The Powassan virus, a flavivirus that is spread by the bite of infected ticks,1 occurs in Ontario and the eastern United States, and produces a nonsuppurative, focal necrotizing meningoencephalitis in horses. Approximately 13% of horses sampled in Ontario in 1983 were serologically positive to the virus. Experimental intracerebral inoculation of the Powassan virus into horses resulted in a neurologic syndrome within 8 days. Clinical findings include a “tucked-up” abdomen, tremors of the head and neck, slobbering and chewing movements resulting in foamy saliva, stiff gait, staggering, and recumbency. There is a nonsuppurative encephalomyelitis, neuronal necrosis, and focal parenchymal necrosis. The virus has not been isolated from the brain. REFERENCE 1. Dupuis AP II, et al. Parasit Vectors. 2013;6:185.

NIGERIAN EQUINE ENCEPHALITIS Nigerian equine encephalitis, a disease with low morbidity but high mortality, is characterized by fever, generalized muscle spasms,

ataxia, and lateral recumbency of 3 to 5 days’ duration. The virus has not been identified, but the only report describes the lesions as consistent with an alphavirus, although Lagos bat virus, a pathogenic lyssavirus, is highly endemic in this area.

MAIN DRAIN VIRUS ENCEPHALITIS The main drain virus has been isolated from a horse with severe encephalitis in California.1 Clinical findings included incoordination, ataxia, stiffness of the neck, head-pressing, inability to swallow, fever, and tachycardia. The virus is transmitted by rabbits and rodents and by its natural vector, Culicoides variipennis. REFERENCE

1. Wilson WC, et al. Rev - Off Int Epizoot. 2015;34:419.

BORNA DISEASE Borna disease is an infectious encephalomyelitis of horses and sheep first recorded in Germany. It is associated with a negative sense, single-stranded RNA virus classified as Bornavirus within the order Mononegavirales. There is a recently recognized avian variant of Borna disease virus, which causes disease in birds.1 The disease and the virus in horses are indistinguishable from EEE. Borna disease is now recognized as a subacute meningoencephalitis in horses, cattle, sheep, rabbits, and cats in Germany, Sweden, and Switzerland.2 There are reports of encephalitis with Borna disease virus genome detected in lesions by PCR in a horse and a cow in Japan. The disease apparently occurs in New World camelids.3 Encephalitis associated with Borna disease virus was detected in young ostriches in Israel. The disease does not appear to be a common cause of nonsuppurative encephalitis in pigs.4 Serologic evidence of infection by Borna disease virus is widespread both geographically and in the range of species.5,6 Borna disease virus is suspected of causing disease in humans, including lymphocytic meningoencephalitis, but infection is not associated with an increased prevalence of psychiatric disorders. Others suggest that the presence of circulating Borna disease virus immune complexes (Borna disease virus antigen and specific antibodies) is associated with severe mood disorders in humans. The role, if any, of Borna disease virus in human neurologic or psychiatric disease has not been established with any certainty and is the subject of considerable debate.1 Detection of Borna disease virus genome by PCR analysis suggests that, although the spontaneous disease in horses and sheep occurs predominantly if not exclusively in Europe, clinically unapparent Borna disease

Viral Diseases Primarily Affecting the Cerebrum

VetBooks.ir

virus infection is widespread in a number of species including horses, cattle, sheep, cats, and foxes. However, concern has been raised that some of these reports might be based on flawed laboratory results as a consequence of contamination of PCR assays. Antibodies to Borna disease virus in serum or CSF have been detected in horses in the eastern United States, Japan, Iran, Turkey, France, and China, and in healthy sheep and dairy cattle in Japan. In areas in which the disease is not endemic, between 3% (United States) and 42% (Iran) of horses have either antibodies or Borna disease virus nucleic acid, detected by PCR, in blood or serum. Similarly, approximately 12% to 20% of horses have serologic evidence of exposure to Borna disease virus in areas of Europe in which the disease is endemic. Antibodies to Borna disease virus and nucleic acid have been detected in humans in North America, Europe, and Japan. Closed flocks of sheep and herds of horses have evidence of persistent infection of some animals, based on serologic testing. It is worth noting that animals infected with the virus and those who are clinically ill may have undetectable to very low antibody titers. The method of transmission of infection between animals is unknown, but it is thought to be horizontal by inhalation or ingestion. Seropositive, clinically normal horses and sheep can excrete virus in conjunctival fluid, nasal secretions, and saliva, suggesting that they might be important in the transmission of infection. Removal of all seropositive and Borna disease virus RNApositive sheep from a closed flock did not prevent seroconversion of other animals in the flock the following year. The possibility of vertical transmission is raised by the finding of Borna disease virus RNA in the brain of a fetal foal of a mare that died of Borna disease. There is a seasonal distribution to the prevalence of the disease, with most cases in horses occurring in spring and early summer. The virus has not been isolated from arthropods, including hematophagous insects. The morbidity in Borna disease is not high, approximately 0.006% to 0.23% of horses affected per year in endemic areas of Germany, but most affected animals die. The pathogenesis of the disease involves infection of cells of the CNS. It is assumed that the virus gains entry to the CNS through trigeminal and olfactory nerves, with subsequent dissemination of infection throughout the brain. Viral transcription and replication occurs within the cell nucleus. Viral replication does not appear to result in damage to the infected neuron. However, infected cells express viral antigens on their surface, which then initiate a cell-mediated immune response by the host that then destroys infected cells (immunosuppression prevents development of the disease). The inflammatory response is largely composed of CD3

lymphocytes. The disease is subacute; infection and the development of lesions may take weeks to months. Clinically inapparent infection appears to be common in a number of species, including horses. In field outbreaks the incubation period is about 4 weeks and possibly up to 6 months. Clinical signs of the disease in horses include the following: • Moderate fever • Pharyngeal paralysis • Lack of food intake • Muscle tremor • Defects in proprioception • Hyperesthesia • Blindness or visual defects7 Lethargy, somnolence, and flaccid paralysis are seen in the terminal stages, and death occurs 1 to 3 weeks after the first appearance of clinical signs. Infection without detectable clinical signs is thought to be common on infected premises. The frequency with which Borna disease virus is detected in horses with gait deficits is greater than in clinically normal horses, suggesting a role for the virus in inducing subtle disease. The presentation of the disease in cattle is similar to that in horses, with affected animals having reduced appetite, ataxia, paresis, and compulsive circling. The disease ends in the death of the animal after a 1- to 6-week course. Hematology and routine serum biochemistry are typically normal, with the exception of fasting-induced hyperbilirubinemia in anorexic horses. Clinicopathologic identification of exposed animals is achieved with complement fixation, ELISA, Western blot, or indirect immunofluorescent tests. At necropsy there are no gross findings, but histologically there is a lymphocytic and plasmacytic meningoencephalitis, affecting chiefly the brainstem, and a lesser degree of myelitis. The highest concentration of virus is in the hippocampus and thalamus. The diagnostic microscopic finding is the presence of intranuclear inclusion bodies within neurons, especially in the hippocampus and olfactory bulbs. The virus can be grown on tissue culture and demonstrated within tissues by immunofluorescence and immunoperoxidase techniques. Borna disease virus can also be detected in formalin-fixed, paraffin-embedded brain tissues using a nested PCR. Specific control measures cannot be recommended because of the lack of knowledge of means of transmission of the virus. The role of inapparently infected horses in transmission of the disease is unknown, and there is no widespread program for testing for such horses. An attenuated virus vaccine was produced by continued passage of the virus through rabbits and used in the former East Germany until 1992. However, its use was discontinued because of questionable efficacy.

1283

FURTHER READING Lipkin WI, et al. Borna disease virus—Fact and fantasy. Virus Res. 2011;162:162-172.

REFERENCES

1. Lipkin WI, et al. Virus Res. 2011;162:162. 2. Lutz H, et al. J Feline Med Surg. 2015;17:614. 3. Jacobsen B, et al. J Comp Pathol. 2010;143:203. 4. Bukovsky C, et al. Vet Rec. 2007;161:552. 5. Bjornsdottir S, et al. Acta Vet Scand. 2013;55:77. 6. Kinnunen PM, et al. J Clin Virol. 2007;38:64. 7. Dietzel J, et al. Vet Pathol. 2007;44:57.

TESCHOVIRUS INFECTIONS Important enteric viruses of the pig belong to the Picornaviridae particularly enteroviruses, teschoviruses and sapeloviruses (formerly porcine enterovirus A or porcine enterovirus).

SEROTYPES

The most important disease of this group is Teschen itself, which was restricted to a particular region around the town of Teschen in Czechoslovakia and the surrounding parts of Eastern Europe.1,2 The mild forms of the disease have occurred elsewhere and are referred to as Talfan or in the past poliomyelitis suum or benign enzootic paresis, and these are probably present worldwide.

SYNOPSIS Etiology Porcine enteroviruses capable of causing encephalomyelitis. Teschen virus, Talfan virus, and others. Epidemiology Certain European countries, Scandinavia, and North America. Morbidity 50%; case fatality 70%–90%. Teschen in Europe. Talfan in UK. Viral encephalomyelitis in North America. Transmitted by direct contact. Signs Acute Teschen: fever, stiffness, unable to stand, tremors, convulsions, and death in few days Subacute Talfan: milder than acute form. Most common in pigs under 2 weeks of age. Morbidity and case–fatality rate 100%. Outbreaks. Hyperesthesia, tremors, knuckling of fetlocks, dog-sitting, convulsions, blindness, and death in a few days. Milder in older growing pigs and adults. Clinical pathology Virus-neutralization tests. Lesions Nonsuppurative encephalomyelitis. Diagnostic confirmation Demonstrate lesion and identify virus. Differential diagnosis list • Pseudorabies • Hemagglutinating encephalomyelitis virus Treatment None. Control Outbreaks will cease and herd immunity develops.

1284

Chapter 14 

■ 

Diseases of the Nervous System

ETIOLOGY

VetBooks.ir

Originally, there were at least 13 enterovirus members, and these are now reclassified. The viruses are resistant to environmental effects (in one study of disinfectants only sodium hypochlorite was effective), are stable, and easily cultivated. The only known host is the pig, and the viruses are not zoonotic. Important enteric viruses belong to the Picornaviridae and the genera Enterovirus, Teschovirus, and Sapelovirus (these were formerly known as porcine enterovirus A or porcine enterovirus serotype B.1 In a survey of 206 viral isolates 97 (47%) were identified as teschoviruses, 18% as sapeloviruses, and 3% as adenoviruses.3 Porcine enteric picornaviruses produce asymptomatic infections as well as reproductive disorders, diarrhea, pneumonia, and dermal lesions. These viruses were previously classified as enteroviruses. They are now reclassified into three groups on the basis of genomic sequences: (1) porcine teschoviruses (PTVs) with 11 different serogroups; (2) porcine enterovirus B, which corresponds to the former enterovirus serotypes 9 and 10; and (3) porcine sapelovirus (PSV), which corresponds to former enterovirus type 8 and has a single serotype that is divided into antigenic variants (PEV 8a, 8b, and 9c). It is associated with reproductive disease, diarrhea, and pneumonia. It appears that PTV-1, the most virulent type, is only found in Central Europe (there have been a number of independent isolates, such as the Konratice and Reporyje strains) and Africa. Talfan virus, isolated from England, and other unnamed isolates appear less virulent. Teschen and Talfan virus occur in subgroup 1, which is now called porcine enterovirus group 1 (PEV-1), but isolates from encephalomyelitis are also associated with other subgroups. The other PTVs and PSV are ubiquitous. Porcine enterovirus B (PEV-9 and PEV-10) is found in Italy, UK, and Japan.4 A PTV caused respiratory distress and acute diarrhea in China in 50-to 70-day-old pigs.5 PTV-8 (a sapelovirus in the new classification) caused a SMEDI-like syndrome in China,6,7 in which approximately 80 gilts aborted and many piglets were stillborn or died soon after birth; samples from most were PTV positive. Within subgroups, strains may be further differentiated using a complement fixation test and monospecific sera. There is variation in virulence between strains, and with many strains, clinical encephalitis following infection appears to be the exception rather than the rule. Most of the infections are subclinical. Polioencephalomyelitis is associated with PTV-1, 2, 3, and 5; reproductive disease is associated with PTV-1, 3, and 6; diarrhea is associated with PTV-1, 2, 3, and 5; pneumonia is associated with PTV-1, 2, and 3; pericarditis and myocarditis have been associated

with PTV-2 and 3; and cutaneous lesions are associated with PTV-9 and 10.

EPIDEMIOLOGY Occurrence and Prevalence of Infection There is serologic evidence that the disease occurs throughout the world. The most severe form of the disease, Teschen disease, appears to be limited to Europe and Madagascar, but the milder forms occur extensively in Europe (Hungary, 2012), Scandinavia, and North America (2002–2007) and recently in Japan (2012). The recent outbreak in the United States (Indiana) was ascribed to porcine enterovirus Serogroup 5 or 6 with the only characteristic feature being the histologic lesions of polioencephalomyelitis. Losses caused by the disease result primarily from deaths. Serologic surveys in areas where the disease occurs indicate that a high proportion of the pig population is infected without any clinical evidence of the disease. In the majority of field occurrences, porcine encephalomyelitis is a sporadic disease affecting either one or a few litters, or a small number of weaned pigs. Morbidity and Case Fatality The morbidity rate is usually about 50% and the case–fatality rate 70% to 90% in Teschen. Talfan is much milder, and the morbidity rate below 6%. Methods of Transmission Infection is transmitted by the fecal–oral route and therefore by ingestion and possibly by aerosol. The virus replicates primarily in the intestinal tract, particularly the lower intestine and the ileum but also in the respiratory tract. Replication is thought to be in the reticuloendothelial cells of the lamina propria. There may be a viremia in the Teschen type of disease but not in the mild forms. Piglets may pick up the infection after weaning when the maternal antibody disappears. Many strains can infect the pig. They can be infected at any age with a strain that they have not been exposed to before. When infection first gains access to a herd, the spread is rapid and all ages of pigs may excrete virus in their feces. Risk Factors Animal Risk Factors Depending on the virulence of the infecting strain, clinical disease primarily affects young pigs but may occur in older pigs at the same stage. As infection becomes endemic and herd immunity develops, excretion of the virus is largely restricted to weaned and early grower pigs. Adults generally have high levels of serum antibody, and suckling piglets are generally protected from infection by colostral and milk antibody. Sporadic disease in suckling pigs may occur in these circumstances in the litters of nonimmune or

low-antibody sows, and may also occur in weaned pigs as they become susceptible to infection. In the recent outbreak in the United States, the major factor was the rapid decline of the maternal antibody in the piglets ( 1, then the agent can persist indefinitely; initial estimates for

1289

R0 before the first feed ban in 1988 ranged from 10 to 12. This degree of infectivity was consistent with the potential that a maximally infectious animal could infect up to 400 other cattle. Since the feed ban, the value for R0 is thought to have decreased to 0 to 0.25, indicating that the disease will soon disappear. Risk for Occurrence of Disease in Countries Changes in the method of processing MBM have occurred in countries other than the UK, and scrapie occurs in sheep in other countries. However, the major risk for the occurrence of the disease in other countries is the importation of latently infected cattle and/or the importation of infected MBM. This risk can be substantially avoided by prohibiting the feeding of MBM to cattle. An assessment in 1996 of risk for the occurrence of BSE in the United States concluded that the potential risk of an epizootic was small and that there are substantial differences in the strength of the risk factors between the United States and the UK. These result from differences in proportional numbers of sheep and cattle, differences in the nature of the beef and dairy industries, the type of animal used for beef production and the age at slaughter, and differences in the practice of feeding ruminant-derived protein in calf rations, which is uncommon in the United States. Thus the risk of an outbreak similar to that in the UK was considered negligible. However, a case in a native-born cow in the United States occurred in 2005. This, and contemporary cases in Canada suggested that infected MBM was imported to the North American continent at some time, or that in the United States, the case reflected the very low incidence of spontaneous atypical BSE in cattle. The cases in both countries occurred in cattle that were born before the ban on feeding MBM imposed in both countries in 1997. Countries with largely pastoral cattle are at low risk. The International Animal health code of the OIE describes five BSE risk categories for countries based on the importation of cattle from at-risk countries, the importation of potentially infected MBM, the consumption of MBM by cattle and other animals, animal feeding practices, livestock population structure, rendering practices, and the potential for recycling of BSE. In order of increasing incidence of BSE these categories are BSE free, BSE provisionally free, minimal BSE risk, moderate BSE risk, and high BSE risk. Experimental Reproduction Although studies on the transmissibility and experimental reproduction of BSE were established before the occurrence of human cases of BSE (vCJD), they have been critical in determining the risk of cattle products

1290

Chapter 14 

■ 

Diseases of the Nervous System

VetBooks.ir

for human disease and the risk for disease in other species. In cattle, disease has been experimentally reproduced by oral and intracerebral inoculation with infected cattle brain homogenates. Oral, intravenous, and intracerebral inoculation of sheep with infected cattle brain homogenates also results in disease. Disease has also been reproduced in goats and mink by parenteral challenge. In pigs, disease has been produced by intracerebral challenge with infected brain homogenates but not oral challenge. It has not been produced by any route of challenge in poultry and is not produced by oral challenge in farmed deer. Infectivity of Tissues Brain, spinal cord, and retina are tissues that are infective to cattle or laboratory animals from natural cases of BSE. The tissues that are infective to cattle or laboratory animals from experimentally infected cattle are brain, spinal cord, retina, distal ileum, bone marrow, trigeminal nerve, and lingual lymph tissue. The infective dose of brain material from a cow with classical BSE appears to be 1000 ppm had blood thiamine levels lower than those drinking water with low levels 40%

>80%

25%

25%

>90%

Variable

Mortality among affected pigs

Medium to high

Low

High

High

High

Variable

Sex of affected pigs

Both

Both

Male

Both

Both

Any

Breed of dam (pure or crossbred)

Any

Any

Landrace

Saddleback

Any

Any

Recurrence in successive litters of same parents

No

No

Yes

Yes

Yes

?

Duration of outbreak

1 year of age) of clinical signs that are

Eye and Conjunctival Diseases

VetBooks.ir

Fig. 16-12  Advanced case of bilateral convergent strabismus with exophthalmos in a German Brown cow. (Reproduced, with permission, from Mömke S, Distl O. Bilateral convergent strabismus with exophthalmus [BCSE] in cattle. An overview of clinical signs and genetic traits. Vet J 2007; 173:272-277.7)

progressive, including bilateral, symmetric, permanent rotation of the eyeballs in an anterior-medial direction and slight to severe laterodorsal exophthalmos (Fig. 16-12).7 Parts of the lateral rectus muscle or retrobulbar fat pad can become visible in severely affected animals. Epiphora is common in cattle with advanced BCSE. The sclera becomes darkly dark pigmented. Mildly affected animals compensate well and can be difficult to detect without close examination of the eyes, whereas more severely affected animals clearly have visual impairment up to and including blindness. There is no effective treatment. An inherited, congenital corneal opacity occurs in Holstein cattle. The cornea is a cloudy blue color at birth, and both eyes are equally affected. Although the sight of affected animals is restricted they are not completely blind, and there are no other abnormalities of the orbit or the eyelids. Histologically there is edema and disruption of the corneal lamellae. Lens dystrophy occurs in Brown Swiss cattle that are affected by an inherited congenital blindness. Japanese Black cattle also suffer from an inherited blindness caused by defects in the pupil, retina, and optic disk. Congenital cataracts occur in a variety of breeds of cattle, and some have a suspected genetic component.10 Multiple cataracts in a herd of Ayrshire cattle in Ireland were not clearly inherited, but the cause was not determined.10 The condition of bilateral cataracts has been observed to be an inherited defect in Romney sheep. It is inherited as an autosomal-dominant trait and can be eradicated easily by culling. Congenital cataracts in Exmoor ponies in Canada are

inherited in a sex-linked fashion, with the disease being significantly more common in females.11 Complete absence of the iris (aniridia) in both eyes is also recorded as an inherited defect in Belgian horses. Affected foals develop secondary cataracts at about 2 months of age. Total absence of the retina in foals has also been recorded as being inherited in a recessive manner. Congenital stationary night blindness (CSNB) in Appaloosa horses is associated with homozygosity for the gene conferring the coat spotting pattern in horses, which itself is caused by a single incomplete dominant gene (LP).5,12 LP maps to a 6-cM region on ECAl. Expression of transient receptor potential cation channel, subfamily m, member 1 (TRPM1) in the retina of homozygous Appaloosa horses is 0.05% the level found in non-Appaloosa horses. Decreased expression of TRPM1 in the eye and the skin may alter bipolar cell signaling and melanocyte function, thus causing both CSNB and LP in horses.5 Microphthalmia is reported to be an inherited defect in Texel sheep, but the incidence is low. It is a well-recognized genetic defect of Texel sheep in Europe. Following importation and “breeding up” of the breed in New Zealand in the 1990s, animals were released from quarantine for further expansion of the breed. The abnormality has occurred in a number of flocks in New Zealand, and an experimental breeding flock is maintained to study the molecular genetics. It is inherited as an autosomal-recessive trait. An outbreak in Texel sheep in New Zealand has been recorded. The optic globes are approximately one-half normal size, and the optic nerves at the chiasma are approximately one-half normal size. No other lesions are present in any organs. The retina is composed of an irregular mass attached to and continuous with the ciliary apparatus at one pole and connected to the optic nerve posteriorly by a short stalk. The morphology and morphogenesis of the defect has been followed in embryos at different ages from ewes known to be carriers of the microphthalmia factor. The primary event was abnormal development of the lens vesicle, with disintegration of the lens and subsequent overgrowth of mesenchymal tissue. The mesenchymal tissue later differentiated in various directions, whereas the epithelial structures found in the microphthalmic eyes at days 56 and 132 of gestation and in newborn lambs appeared to be remnants of the epithelial lens vesicle. Typical colobomata, ophthalmoscopically visible defects of one or more structures of the eye, caused by an absence of tissue, have assumed a more prominent position than previously because of their high level of occurrence in Charolais cattle. The lesions are present at birth and do not progress beyond that stage. They affect vision very

1659

little, if at all. However, because they are defects they should be named in certificates of health, but they are not usually considered as being a reason for disqualification from breeding programs. In Charolais cattle the inheritance of the defect is via an autosomaldominant gene with complete penetrance in males and partial (52%) penetrance in females. The prevalence may be as high as 6%, and in most cases both eyes are affected. The defect is a result of incomplete closure of one of the ocular structures at or near the line of the embryonic choroidal fissure. Failure of the fissure to close represents the beginnings of the coloboma. The retina, choroid, and sclera are usually all involved. Entropion is inherited in a number of sheep breeds, including Oxfords, Hampshires, and Suffolks. Affected lambs are not observed until about 3 weeks of age when attention is drawn to the eyelids of the apparent conjunctivitis. A temporary blindness results, but even without treatment there is a marked improvement in the eyelids. Congenital entropion occurs in related Boer goat kids, but the mode of inheritance, if any, is unknown.13 Distichiasis, in which aberrant cilia are present at the eyelid margin, appears to occur with greater frequency in Friesian horses, in which rigid cilia cause corneal irritation or corneal ulceration. Although an inherited cause is suspected, the etiology is unclear.14 Ocular dermoids are recorded as genetically transmitted in Hereford cattle. They occur as multiple small masses of dystrophic skin complete with hair on the conjunctiva of both eyes of affected cattle. They can be anywhere on the cornea, on the third eyelid, or the eyelid, and they may completely replace the cornea; there may be a resulting marked dysplasia of the internal ocular structures. Ocular dermoid cysts are single, solid, skin-like masses of tissue, adherent usually to the anterior surface of the eye, causing irritation and interfering with vision (Fig. 16-13). The eyelid, the third eyelid, and the canthus may also be involved, and the lesions may be unilateral or bilateral. When they occur at a high frequency in a population, it is likely they are inherited, as they can be in Hereford cattle. It is also recorded in foals. The defect is sometimes associated with microphthalmos. Surgical ablation is recommended. Nasolacrimal duct fistulae, either unilateral or bilateral, occur in Brown Swiss cattle. The defect, evidenced by persistent epiphora and presence of a nasolacrimal fistula medial to the medial canthus of the eye, is inherited, although the mode of inheritance is unclear.15,16 Combined Ocular Defects Although the vision appears unaffected, a large number of congenital defects of the eye

1660

Chapter 16 

■ 

Diseases of the Skin, Eye, Conjunctiva, and External Ear

VetBooks.ir

high-frequency (approximately 100 to 200 horizontal oscillations/minute) nystagmus of both eyes, with the eyes moving approximately 1 mm. Nystagmus has been observed shortly after birth in some calves, but the age of onset is not accurately known. Adult animals appear to be unaffected by the nystagmus and appear to have normal vision and balance and ocular reflexes. Pendular nystagmus is not thought to affect production and should not be mistaken as indicating the presence of a serious neurologic disease. REFERENCES

Fig. 16-13  Ocular dermoid cyst on the ventral corneal and limbus of the left eye of a Simmental calf.

have been observed in cattle, including Herefords, affected by partial albinism. The defects include iridal heterochromia, tapetum fibrosum, and colobomas. Congenital blindness is also seen in cattle with white coat color, especially Shorthorns. The lesions are multiple and include retinal detachment, cataract, microphthalmia, persistent pupillary membrane, and vitreous hemorrhage. Internal hydrocephalus is present in some, and hypoplasia of optic nerves also occurs. A combination of iridal hypoplasia, limbic dermoids, and cataracts occurred in the progeny of a Quarter horse stallion, presumably as a result of a mutation in the stallion and transmission to the foals via an autosomal-dominant gene. The inheritance is simple autosomal recessive. Irideremia (total or partial absence of iris), microphakia (smallness of the lens), ectopia lentis, and cataract have been reported to occur together in Jersey calves. The mode of inheritance of the characters is as a simple recessive trait. The calves are almost completely blind but are normal in other respects and can be reared satisfactorily if they are hand-fed. Although the condition has been recorded only in Jerseys, similar defects, possibly inherited, have also been seen in Holsteins and Shorthorns. Multiple congenital ocular abnormalities occur with high frequency in Rocky Mountain horses and the closely related breeds Kentucky Mountain Saddle horse and Mountain Pleasure horse.1,12,17,18 The cause is a missense mutation in the PMEL gene that has pleiotropic effects on the eye and coat color, causing a dilute or “silver” coat.17 Similar to the silver mutation, MCOA is controlled by a dominant gene, with some

reports demonstrating a codominant mode of inheritance and incomplete penetrance.3,18 Homozygotes are thought to be more severely affected, having multiple abnormalities, whereas heterozygotes have cysts only, although this may not always be the case. Incomplete penetrance of this disorder has made studying the molecular mechanism behind these eye phenotypes difficult. Individuals carrying the causative mutation that are phenotyped as normal may either have cysts that were too small to detect or be true cases of nonpenetrance.18 Equine MCOA is characterized by a diverse set of ocular phenotypes.18 The predominant phenotype consists of large cysts, which are often bilateral, originating from the temporal ciliary body or peripheral retina, and additional phenotypes include abnormalities of the cornea, iris, lens, and iridocorneal angle.18

INHERITED NYSTAGMUS Familial Undulatory Nystagmus Familial undulatory nystagmus is an inherited defect of Finnish Ayrshire cattle characterized by a tremor-like, synchronous movement of the eyeballs. The tremor has small amplitude (1 to 2 mm) and fast (200/ min) rate and is usually vertical. Nystagmus is present at all times, there is no sign of impaired vision, and the eye reflexes are normal. The condition is a blemish rather than a disease because there is no functional deficiency. Pendular Nystagmus Pendular nystagmus is an inherited defect of Holstein–Friesian cattle observed primarily in North America. A report from 1981 utilizing a convenience sample in New York state reported a prevalence of 0.51% in 2932 cattle from 62 herds. Affected cattle have a

1. Kaps S, et al. Pferdeheilkunde. 2010;26:536. 2. Tetens J, et al. Tierarztl Prax Ausg G Grosstiere Nutztiere. 2007;35:211. 3. Andersson LS, et al. BMC Genet. 2008;9. 4. Andersson LS, et al. Mamm Genome. 2011;22: 353. 5. Bellone RR, et al. Genetics. 2008;179:1861. 6. Brunberg E, et al. BMC Genet. 2006;7. 7. Moemke S, et al. Vet J. 2007;173:272. 8. Fink S, et al. Mol Vision. 2012;18:2229. 9. Momke S, et al. Anim Gen. 2008;39:544. 10. Krump L, et al. Irish Vet J. 2014;67. 11. Pinard CL, et al. Vet Ophthalmol. 2011;14:100. 12. Bellone RR. Anim Gen. 2010;41:100. 13. Donnelly KS, et al. Vet Ophthalmol. 2014;17:443. 14. Hermans H, et al. Equine Vet J. 2014;46:458. 15. Braun U, et al. BMC Vet Res. 2014;10. 16. Braun U, et al. Schweiz Arch Tierheilkd. 2012;154:121. 17. Andersson LS, et al. PLoS ONE. 2013;8. 18. Grahn BH, et al. Can Vet J. 2008;49:675.

External Ear Diseases OTITIS EXTERNA Otitis externa, inflammation of the skin and external auditory canal, can affect cattle of all ages, in isolated cases, an entire herd, or in entire regions. Arthropod parasites, foreign bodies, and sporadic miscellaneous infections may cause irritation in the ear, accompanied by rubbing of the head against objects and frequent head-shaking. In tropical and subtropical regions, parasitic otitis is more important than in other more temperate regions. The mites Raillietia auris and Dermanyssus avium, the tick Otobius magnini, larvae (Stephanofilaria zahaeeri), free-living nematodes (Rhabditis bovis), and the blue fly (Chrysomia bezziano) are of importance in Europe, Africa, India, and America. Malassezia spp., Candida spp., Rhodotorula mucilaginosa, Aspergillus spp., and Micelia sterilia are common causes of otitis externa in cattle in Brazil. When the syndrome occurs in a large number of animals in a herd, as it does in tropical countries, it is necessary to identify the specific causative agent. R. bovis is a common cause. Affected animals are depressed, eat little, and appear to experience

External Ear Diseases

VetBooks.ir

pain when they swallow, and they shake their heads frequently. Both ears are affected in most cases, and there is a stinky, bloodstained discharge that creates a patch of alopecia below the ear. The area is painful when touched, the external meatus of the aural canal is obviously inflamed, and the parotid lymph nodes are enlarged. Extension to the middle ear is an unusual sequel. Topical treatment with ivermectin and a broadspectrum antibiotic is effective. Circumscribed ulcerative lesions on the ears with raised edges frequently associated with secondary bacterial or fungal infection are a common finding in cattle and buffaloes affected by buffalopox virus infection (see also “Coxpox and Bufallopox”).

and it was suggested that the condition is multifactorial.1

FURTHER READING

CLINICAL SIGNS

Duarte ER, Hamdan JS. Otitis in cattle, an aetiological review. J Vet Med B Infect Dis Vet Public Health. 2004;51:1-7.

EAR-TIP NECROSIS Currently, ear-tip necrosis of pigs appears to be a more common condition.

ETIOLOGY

The condition may be associated with the presence of Treponema pedis. It can be cultured from the lesions and from the gingivae of pigs. It is anaerobic, fastidious, 4 mm to 6 mm in length, and 0.25 microns in diameter. There may be a sequence of infections when Staphylococcus hyicus is followed by the spirochetes and then infected with streptococci. In a recent study of putative agents, no single cause could be found,

EPIDEMIOLOGY

Ear-tip necrosis is usually seen in pigs at 1 to 16 weeks of age with a peak around 8 to 10 weeks. It may also occur in older pigs, when it is usually seen at the base of the ear. Typically it may occur in only one litter of pigs, and 80% may be affected. It may be associated with mixing and moving when a lot of pigs show ear biting. Contributing factors are thought to include poor hygiene, high humidity, low air changes, overstocking, abrasions on feeders and pen divisions, and fighting associated with moving and mixing. The affected pigs appear to show little evidence of distress and often recover spontaneously, and in these cases the only evidence of the condition is a crinkled edge to the ears. When it first appears, if the grease on the ear is removed you can see a crack in the skin, which obviously then allows bacterial penetration. Some persistent lesions may enlarge and spread. Occasionally, pigs show inappetence, unthriftiness, fever, or even death, often as a result of secondary infections.

PATHOLOGY

The lesions are black areas of necrosis with ulcers on the tips of the ears and the caudal edge of the ears. The lesions are dry and crusty, and in some cases there may be loss of the whole ear or part of the ear. This is caused by progressive thrombi formation

1661

leading to ischemia because there is a poor collateral circulation in the ear. In cases reported in Sweden, spirochetes were observed in silver-stained histologic sections, and a spirochete isolate was obtained and identified as a yet unnamed species of the genus Treponema closely resembling those found in digital dermatitis in cattle. The same organism was isolated from oral samples, along with T. socranskii.2

DIFFERENTIAL DIAGNOSIS

Simple ear biting is the main differential, but this usually starts at the base of the ear. Other septicemic causes of ear tissue loss, such as H. parasuis, Salmonella, or S. suis, may be suspected when ears are discolored, congested, or necrotic.

TREATMENT

Antibiotic sprays may or may not help.2,3 A recent study has suggested that vaccination for PCV-2 infections may reduce the incidence of ear-tip necrosis.4 REFERENCES

1. Weissenbacher-Lang C, et al. Vet J. 2012;194:392. 2. Pringle M, et al. Vet Micro. 2010;139:279. 3. Pringle M, et al. Vet Micro. 2010;142:461. 4. Pejsak Z, et al. Res Vet Sci. 2011;91:125.

INHERITED CROP EARS Inherited as a single autosomal-dominant incomplete character in Bavarian Highland cattle, the crop ear anomaly affects both ears, appears at birth, and varies from a minor trimming up to a complete deformity and reduction in size.

VetBooks.ir

17 

Metabolic and Endocrine Diseases

INTRODUCTION  1662 METABOLIC DISEASES OF RUMINANTS  1662 Periparturient Period in Cattle and Sheep  1662 Metabolic Profile Testing  1667 Parturient Paresis (Milk Fever)  1675 Acute Hypokalemia in Cattle  1690 Downer-Cow Syndrome  1693 Hypomagnesemic Tetanies  1699 Hypomagnesemic Tetany (Lactation Tetany, Grass Tetany, Grass Staggers, Wheat Pasture Poisoning)  1699 Hypomagnesemic Tetany of Calves  1706 Transport Recumbency of Ruminants  1707 Ketosis and Subclinical Ketosis (Hyperketonemia) in Cattle  1708 Fatty Liver in Cattle (Fat-Mobilization Syndrome, Fat-Cow Syndrome, Hepatic Lipidosis, Pregnancy Toxemia in Cattle)  1716 Pregnancy Toxemia (Twin Lamb Disease) in Sheep  1722 Steatitis, Panniculitis, and Fat Necrosis  1726

Introduction Metabolic diseases are very important in dairy cows and pregnant ewes. In the other livestock species, metabolic diseases occur only sporadically. The high-producing dairy cow always verges on abnormal homeostasis, and the breeding and feeding of dairy cattle for high milk yields is etiologically related to metabolic disease so common in these animals. The salient features of the common metabolic diseases of farm animals are summarized in Table 17-1. The term production disease includes those diseases previously known as metabolic diseases, such as parturient paresis (milk fever), hypokalemia, hypomagnesemia, hyperketonemia and ketosis, hyperlipemia, and other conditions that are attributable to an imbalance between the rates of input of dietary nutrients and the output of production. When the imbalance is maintained, it may lead to a change in the amount of the body’s reserves of certain metabolites and their “throughput.” This generalization applies principally to energy balance (such as ketosis and hypoglycemia), in addition to hypomagnesemia, and to a lesser extent hypocalcemia. In these diseases output is greater than input, either because of the Copyright © 2017 Elsevier Ltd. All Rights Reserved. 1662

INHERITED METABOLIC DISEASES OF RUMINANTS  1727 Deficiency of UMP Synthase (Dumps)  1727 Hepatic Lipodystrophy in Galloway Calves  1727 METABOLIC DISEASES OF HORSES  1727 Equine Pituitary Pars Intermedia Dysfunction (Formerly Equine Cushing’s Disease)  1727 Equine Metabolic Syndrome  1731 Pheochromocytoma (Paraganglioma)  1736 Glycogen Branching Enzyme Deficiency in Horses  1736 Lactation Tetany of Mares (Eclampsia, Transport Tetany)  1736 Equine Hyperlipemia  1737 DISORDERS OF THYROID FUNCTION (HYPOTHYROIDISM, HYPERTHYROIDISM, CONGENITAL HYPOTHYROIDISM, THYROID ADENOMA)  1739 Iodine Deficiency  1742 Inherited Goiter  1747

DISEASES CAUSED BY NUTRITIONAL DEFICIENCIES  1747 Introduction  1747 Evidence of a Deficiency as the Cause of the Disease  1747 Evidence of a Deficiency Associated With the Disease  1748 Evidence Based on Cure or Prevention by Correction of the Deficiency  1748 DEFICIENCIES OF ENERGY AND PROTEIN  1753 Deficiency of Energy  1753 Deficiency of Protein  1753 Low-Milk-Fat Syndrome  1754 DISEASES ASSOCIATED WITH DEFICIENCIES OF MINERAL NUTRIENTS  1754 Prevalence and Economic Importance  1754 Diagnostic Strategies  1754 Deficiencies in Developing Countries  1755 Pathophysiology of Trace-Element Deficiency  1755 Laboratory Diagnosis of Mineral Deficiencies  1756

selection of cattle that produce so heavily that no naturally occurring diet can maintain the cow in nutritional balance or because the diet is insufficient in nutrient density or unevenly balanced. For example, a ration may contain sufficient protein for milk production but contains insufficient precursors of glucose to replace the energy excreted in the milk. Although we agree with the generalization on which the term production disease is based, we prefer to continue to use the expression metabolic disease because of common usage and the clinical focus that metabolism must match the level of production.

7-day window starting with parturition has a tremendous influence on morbidity, lactation production, reproductive performance, and mortality. The susceptibility of dairy cows to metabolic disease appears to be related to the extremely high turnover of water, electrolytes, and soluble organic materials during the early part of lactation. With this rapid rate of exchange of water, sodium, calcium, magnesium, chloride, and phosphate, a sudden variation in their excretion or secretion in milk or by other routes, or a sudden variation in their intake because of changes in ingestion, digestion, or absorption, may cause abrupt, damaging changes in the internal environment of the animal. It is the volume of the changes in intake and secretion and the rapidity with which they can occur that affect the metabolic stability of the cow. In addition, if the continued nutritional demands of pregnancy are exacerbated by an inadequate diet in the dry period, the incidence of metabolic disease will increase. The effect of pregnancy is particularly important in ewes, especially those carrying more than one lamb.

Metabolic Diseases of Ruminants PERIPARTURIENT PERIOD IN CATTLE AND SHEEP The incidence of metabolic disease in dairy cattle increases as milk production increases and, in particular, as the rate of increase in milk production increases (called milk yield acceleration). In dairy cows, the total disease incidence rapidly increases in the very late periparturient period, peaks on the day of parturition, and then rapidly declines until day 7 of lactation (Fig. 17-1). This critical

Transition Period in Dairy Cows The transition period is a crucial stage in the production cycle of the dairy cow; no other

Metabolic Diseases of Ruminants

1663

VetBooks.ir

Table 17-1  Salient features of metabolic diseases of farm animals Disease

Etiology and epidemiology

Diagnosis

Treatment

Control

Milk fever of cattle

Hypocalcemia Occurs primarily in dairy cows after third lactation Also in beef cows 48 hours before or after calving and in midlactation

Low serum calcium concentration

Calcium salts IV, SC

Dietary management of anions–cations

Downer cow

Complication of milk fever; recumbent too long before treatment

Clinical findings Serum CK activity

Supportive therapy

Early treatment of milk-fever cases

Acute hypokalemia of cattle

In lactating dairy cows treated with corticosteroids for recurrent ketosis, and mastitis

Low serum potassium concentration

Potassium chloride IV

Avoid excessive use of isoflupredone for recurrent ketosis

Lactation tetany of mares

High-producing lactating mares being nursed by vigorous well-nourished foal a few weeks of age

Low serum calcium concentration

Calcium borogluconate

No reliable method available

Hypomagnesemic tetany (lactation tetany)

Lactating dairy cows on lush fertilized pastures Also in beef cows before and after calving

Low serum magnesium concentration

Magnesium salts IV

Supplementation of diets at strategic times with magnesium salts

Ketosis of cattle

Before and after parturition in cattle

Blood, urine, and milk levels of ketone bodies during the transition period 3 weeks before and after parturition

Glucose IV Propylene glycol and electrolyte solutions orally

Prepartum dietary management of energy intake

Pregnancy toxemia of sheep

Declining plane of nutrition in ewes in late pregnancy

Urinary ketones Hypoglycemia Metabolic acidosis and terminal uremia

Cesarean section or induction of parturition

Nutritional management of pregnant ewes to ensure a rising plane of nutrition in the second half of pregnancy

Fatty liver of cattle

High-producing dairy cows overfed during the dry period In well-conditioned beef cattle in late pregnancy when energy intake suddenly decreased

Ketonemia, ketonuria, hypoglycemia

Poor prognosis in severe cases Fluid and electrolyte therapy, glucose IV, propylene glycol orally and insulin

Nutritional management of pregnant cows to avoid excessive weight gain Avoid situations that reduce feed intake at time of parturition

Equine hyperlipidemia

Deranged fat metabolism Pregnant or lactating middle-aged ponies, donkeys, and American miniature horses worldwide Sporadic

Hyperlipidemia

Enteral or parenteral feeding, insulin, heparin Treat underlying disease

Maintain optimal body condition Prevent disease and nutritional stress in pregnancy

Postparturient hemoglobinuria

Dietary deficiency in high-producing dairy cows 2–4 weeks after calving Copper-deficient area Cruciferous crops

Low serum inorganic phosphorus concentration Low PCV Hemoglobinuria

Whole blood transfusion Sodium acid phosphate IV Dicalcium phosphate orally

Ensure adequate dietary phosphorous intake

period can affect subsequent production, health, and reproductive performance so greatly. The success of the transition period effectively determines the profitability of the cow during that lactation. Nutritional or management limitations during this time may impede the ability of the cow to reach maximal milk production. The primary challenge faced by cows is a sudden and marked increase of nutrient requirements for milk production, at a time when dry matter intake, and thus nutrient supply, lags far behind. Dry matter intake typically declines during the final week before parturition. This decline and changes in endocrine profiles contribute to elevated plasma nonesterified fatty acid (NEFA) concentrations, which have been

related to the occurrence of fat-mobilizationrelated metabolic diseases such as fatty liver and ketosis. The magnitude of the decline in feed intake as parturition approaches may be a better indicator of metabolic health of postpartum cows than the actual level of feed intake. Diet, body-condition score, and parity influence dry matter intake and energy balance. The occurrence of diseases during the transition period results in lost milk production during the time of illness and often for the entire lactation. A key area of the biology of transition cows is lipid metabolism. Excessive lipid metabolism from adipose tissue is linked with a higher incidence of periparturient diseases. Fatty livers were described in ketotic

cows in the 1950s. Hepatic fat accumulation was then noted in normal cows during early lactation. This was followed by a description of a fat-mobilization syndrome in early lactation, in which cows mobilized body lipids from adipose tissue and deposited lipids in the liver, muscle, and other tissues. This was followed by descriptions of elevated nonesterified fatty acid concentrations during the last 7 days before calving being associated with a higher incidence of ketosis, displaced abomasum, and retained fetal membranes, but not associated with the incidence of milk fever. Understanding the metabolism of NEFA by the liver is a critical component of understanding the biology of the transition cow. Extreme rates of lipid

1664

Chapter 17 

■ 

Metabolic and Endocrine Diseases

10 Total disease incidence (cases/100 cows)

8 First lactation Third lactation

7 6 5 4 3 2 1 0 –28

0

28 Days from calving

56

84

0

28 Days from calving

56

84

A 3.0 Milk yield acceleration kg/d/d

VetBooks.ir

9

2.5 2.0 1.5 1.0 0.5 0.0 –0.5 –28

B

Fig. 17-1  A, Total disease incidence (sum of mastitis, ketosis, digestive disorders, and laminitis) relative to days from calving for first- and third-lactation cows in Danish dairy herds that calved in 1998. B, Acceleration in milk yield through lactation for cows peaking at 30 kg or 60 kg (bold line) of milk daily. (Reproduced with permission from Ingvartsen KL. Anim Feed Sci Technol 2006; 126:175-213.)

mobilization lead to increased uptake of NEFA by the liver and an increased rate of triglyceride accumulation in the liver. If this lipid infiltration becomes severe, the syndrome of hepatic lipidosis or fatty liver may result, which can then result in prolonged recovery from other diseases, increased incidence of other diseases, and increased susceptibility to induction of ketosis. During the transition period, dairy cows undergo large metabolic adaptations in glucose, fatty acid, and mineral metabolism. The practical goal of nutritional management during this period is to support these metabolic adaptations. There are two different philosophic approaches for feeding transition cows in animals fed a total mixed ration. The first approach increases the energy density of the diet to “correct” for the anticipated decrease in dry matter intake in late gestation. Increasing the amount of energy supplied through dietary carbohydrate during the prepartum period results in generally positive effects on metabolism and performance of transition cows. In contrast, the second approach focuses on decreasing the energy density and increasing forage (by

the daily provision of 2 to 4 kg of straw) in far-off cattle in an attempt to promote dry matter intake. Attempts to increase energy supply by feeding dietary fat sources or decrease energy expenditure by supplying specific fatty acids such as trans-10, cis12conjugated linoleic acid to decrease milkfat output during early lactation do not decrease the release of NEFAs from adipose tissue. In addition to nutritional management strategies to optimize the health of the transition cow, certain feed additives are in use to reduce subclinical ketosis and reduce the incidence of displaced abomasum. Monensin is a carboxylic polyether ionophore produced by a naturally occurring strain of Streptomyces cinnamonesis. Monensin exerts its many effects by shifting the microbial populations in the rumen; this results in changes in the proportions of short-chain volatile fatty acids in the rumen, specifically increasing propionic acid and reducing the molar percentages of butyric acid and acetic acid. Increased rumen propionic acid concentrations directly lead to increased gluconeogenesis and should therefore decrease the

incidence of ketosis and hyperketonemia in early lactation and improve energy balance. In Canada, monensin is approved to be administered as a controlled-release capsule (CRC) as an aid in the prevention of subclinical ketosis in lactating dairy cattle. The monensin CRC delivers 335 mg of monensin daily for 95 days, improves energy balance, and decreases the incidence of all three energy-associated diseases of lactating dairy cows: retained placenta, displaced abomasum, and clinical ketosis. Cows treated with the monensin CRC at 3 weeks before the anticipated calving date had decreased serum NEFA and β-hydroxybutyrate (BHB) concentrations and increased serum cholesterol and urea concentrations in the week immediately preceding precalving. Monensin has no effect on serum calcium, phosphorus, or glucose concentration in the precalving period. After calving, serum concentrations of BHB and phosphorus concentrations were lower and serum concentrations of cholesterol and urea higher in monensin-treated cows. The lower NEFA values indicate less fat mobilization, and the higher cholesterol suggests greater lipoprotein export from the liver. The higher urea levels are thought to result from a protein-sparing effect in the rumen, resulting in an increased supply of amino acids in the small intestine. There was no effect of treatment on serum NEFA, glucose, or calcium concentrations in the first week postcalving. Daily monensin ingestion starting before calving therefore improves indicators of energy balance in both the immediate precalving and postcalving periods. Voluntary Dry Matter Intake in Periparturient Dairy Cattle The factors affecting voluntary dry matter intake (DMI) of lactating cattle are extremely important and have received much attention for many decades. A substantial decrease in DMI is initiated in late pregnancy and continues into early lactation, with the lowest DMI occurring on the day of calving. Postpartum DMI is considerably higher in multiparous cows compared with primiparous cows and increases after lactation in both groups, but the rate of increases varies widely. In cows given diets of constant composition, the milk yield typically peaks at 5 to 7 weeks postpartum, and the maximum intake is reached between 8 and 22 weeks after calving. The increase in DMI from week 1 postpartum to time of peak intake is affected by the diet fed during lactation and also by prepartum feeding; the latter influences the amount of fat stored and therefore the bodycondition score of the animal. The normal pattern of feed intake may be severely influenced by disease states because both clinical and subclinical infections are known to substantially reduce appetite and performance. The decrease in DMI has traditionally been attributed to physical constraints such

Metabolic Diseases of Ruminants

VetBooks.ir

as the enlarging uterus, but this role may be overemphasized. The decrease in DMI coincides with changes in reproductive status, changes in fat mass, and metabolic changes in support of lactation. A number of metabolic signals may have a role in intake regulation. These signals include nutrients, metabolites, reproductive hormones, stress hormones, leptin, insulin, gut peptides, cytokines and neuropeptides such as neuropeptide Y, galanin, and corticotrophin-releasing factor. Immunosuppression During the Transition Period In addition to the adaptations in classical metabolism, cows during the transition period also undergo a period of reduced immunologic capacity during the periparturient period. The immune dysfunction is broad in scope, affects multiple functions of various cell types, and lasts from approximately 3 weeks before calving until approximately 3 weeks after calving. Cows during this period are more susceptible to mastitis. The etiology of periparturient immunosuppression is multifactorial and not well understood, but it seems to be related to physiologic changes associated with parturition and the initiation of lactation and to metabolic factors related to these events. Glucocorticoids are immunosuppressants, plasma cortisol concentration is increased at parturition, and endogenous glucocorticoids have been postulated to play a role in periparturient immunosuppression. Periparturient cattle have impaired expression of adhesion molecules and decreased migration capacity of blood neutrophils. Because the rapid recruitment of neutrophils into newly infected mammary tissue is the key immunologic defense against mastitis-causing pathogens in ruminants, periparturient neutrophil dysfunction may contribute to the increased susceptibility to mastitis at this time. Metabolic challenges around calving may also play in role in increased susceptibility, as nonesterified fatty acids significantly reduce the in vitro immunosuppressiveness of mononuclear cells of ewes, potentially resulting in impairment of cell-mediated and humoral immunity in sheep and cattle with ketosis. Vitamin E is a fat-soluble membrane antioxidant that enhances the functional efficiency of neutrophils by protecting them from oxidative damage following intracellular killing of ingested bacteria. The parenteral administration of vitamin E has been investigated for the prevention of peripartum diseases such as retained placenta, metritis, and clinical mastitis. Only cows with marginal vitamin E status (serum α-tocopherol < 2.5 × 10–3) 1 week before calving will have a reduction in the risk of retained placenta following a subcutaneous injection of 3000 IU of vitamin E. In cows with an adequate serum vitamin E concentration there was no reduction, and primiparous animals were most

likely to benefit from vitamin E 1 week before parturition. The associations between peripartum serum vitamin E, retinol, and β-carotene concentrations in dairy cattle and disease risk indicated that an increase in α-tocopherol of 1 µg/ML in the last week prepartum reduced the risk of retained placenta by 20%, whereas serum NEFA concentrations ≥ 0.5 mEq/L tended to increase the risk of retained placenta by 80%. In the last week prepartum, a 100-ng/mL increase in serum retinol was associated with a 60%decrease in the risk of early-lactation clinical mastitis. Diseases of Lactation Parturition is followed by the sudden onset of a profuse lactation, which, if the nutrient reserves have already been seriously depleted, may result in clinical metabolic disease. The essential metabolite that is reduced below the critical level determines the clinical syndrome that will occur. Most attention has been paid to variations in balances of calcium and inorganic phosphates relative to parturient paresis, magnesium relative to lactation tetany, and plasma glucose and ketone concentration and hepatic lipidosis relative to ketosis, but it is probable that other imbalances are important in the production of as yet unidentified syndromes. The vast majority of production diseases of dairy cows occur very early in lactation. At this time, the cow is producing milk at a rate that is substantially less than her maximum. In terms of rate, high- and lowmilk-yielding cows are producing rather similar amounts at this time. However, in terms of acceleration, the change in milk yield per day, it is highest immediately after calving. During the succeeding period of lactation, particularly in cows on test schedules and under the strain of producing large quantities of milk, there is often variable food intake, especially when pasture is the sole source of food, and instability of the internal environment inevitably follows. The period of early lactation is an unstable one in all species. Hormonal stimulation at this stage is so strong that nutritional deficiency often does not limit milk production, and a serious drain on reserves of metabolites may occur. Recombinant bovine somatotrophin (rBST, sometribove zinc) is a synthetically derived hormone that may be identical to naturally occurring bovine growth hormone, or slightly modified by the addition of extra amino acids. The product was approved in the United States in 1993, and its use began commercially in 1994 in dairy herds to increase milk production. The product is a sterile, prolonged-release injectable formulation of rBST in single-dose syringes that each contain 500 mg of sometribove zinc. The recommended dosage protocol is one syringe injected subcutaneously (SC) in the postscapular region (behind the shoulders)

1665

or in the ischiorectal fossa (depression on either side of the tailhead) every 14 days beginning during the 9th week after calving and continuing until the end of lactation.2 Approximately 15% of dairy herds in the United States used rBST in 2013. The product has been licensed for use in at least 20 other countries, including Argentina, Brazil, Chile, Colombia, Costa Rica, Ecuador, Egypt, Guatemala, Honduras, Jamaica, Lebanon, Mexico, Panama, Pakistan, Paraguay, Peru, Salvador, South Africa, South Korea, Uruguay, and Venezuela. In comparison, a number of countries, including Australia, Canada, Israel, Japan, New Zealand, and all European Union countries, have not approved its use. A meta-analysis of the effects of rBST on milk production, animal health, reproductive performance, and culling was undertaken. Recombinant bovine somatotrophin was found to increase milk production by 11% in primiparous cows and 15% in multiparous cows, although there was considerable variation in the magnitude of the milk production increase between studies. Some statistically significant effects on milk composition (percentage of butterfat, protein, and lactose) were found; however, they were all very small. Treatment increased dry matter intake by an average of 1.5 kg/d during the treatment period, and dry matter intake remained elevated for the first 60 days of the subsequent lactation. Despite the increase in dry matter intake, treated animals had lower body-condition scores at the end of the treatment period, and the reduced scores persisted until the start of the subsequent lactation. Recombinant bovine somatotrophin increased the risk of clinical mastitis by approximately 25% during the treatment period, but there were insufficient data to draw firm conclusions about the effects of the drug on the prevalence of subclinical intramammary infections as assessed by somatic cell count. The increase in the incidence of clinical mastitis in cattle administered rBST appears similar to that expected from an increase in milk production alone using genetic selection and improved nutrition, milking frequency, and management practices. Use of rBST increased the risk of a cow failing to conceive by approximately 40%. For cows that did conceive, there was no effect on services per conception and only a small increase in average days open. Use of the drug had no effect on gestation length, but the information about a possible effect on twinning was equivocal. Cows treated with rBST had an estimated 55% increase in the risk of developing clinical signs of lameness. There appeared to be an increased risk of culling in multiparous cows. Use of the drug in one lactation period appeared to reduce the risk of metabolic diseases (particularly ketosis) in the early period of the subsequent lactation. It was found that the reproductive effects of the drug could be

1666

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

controlled by delaying its use until the cows were confirmed pregnant. In 1998, an expert panel appointed by the Canadian Veterinary Medical Association at the request of Health Canada found a number of legitimate animal welfare concerns associated with the use of rBST. In 1999 Health Canada announced that it would not approve the use of rBST for sale in Canada on the basis of the health and welfare of cattle. The Royal College of Physicians and Surgeons of Canada’s Expert Panel on Human Safety of rBST found no biologically plausible reason for concern about human safety if rBST were to be approved for sale in Canada. In 1999 a working group from within the Scientific Committee on Animal Health and Animal Welfare of the European Commission presented a more extensive report on rBST that summarized similar results and engaged substantive discussion of animal welfare issues. It concluded that rBST should not be used in dairy cattle in Europe. In October 1999, the European Commission banned the use and marketing of rBST in the European Union as of January 1, 2000. Relationship Between Lactational Performance and Health of Dairy Cattle There is little evidence that high-yielding cows have increased risk of dystocia, retained placenta, metritis, and left-side displacement of the abomasum. The association between high levels of production and periparturient diseases is inconsistent; in general, high levels of management (including nutrition and housing) are usually associated with high levels of production. Although no phenotypical relationship between milk yield and the risk of ketosis and lameness has been found, selection for higher milk yield will probably increase the lactational incidence risk for both diseases. Mastitis is the only disease for which a clear relationship between increased milk yield and increased risk of infection has been found. Continued selection for high milk yield should therefore be expected to increase the incidence of clinical mastitis in dairy cattle. However, some authors have stated that “Reviewing existing literature, even with structured literature selection, is inadequate to the task of elucidating the relationship between the lactational performance and risk of production diseases.”3 The most notable feature of the literature evaluation is the large variability that exists between studies. This strongly suggests that there are important factors that need to be considered before meaningful conclusions concerning the relationship between lactational performance and risk of disease can be drawn. Breed Susceptibility The fact that some dams are affected much more by these variations than others is probably explainable on the basis of variations in

internal metabolism and degree of milk production among species and among individuals. Among groups of cows, variations in susceptibility appear to depend on either genetic or management factors. Certainly, Jersey cows are more susceptible to parturient paresis than cows of other breeds. Even within breeds, considerable variation is evident in susceptibility between families. Under these circumstances, it seems necessary to invoke genetic factors as predisposing causes for metabolic diseases. Management Practices The management practices of most importance are nutrition and housing. In those sections of North America where cattle are housed during the winter and in poor pasture areas, ketosis is prevalent. In the Channel Islands, local cattle are unaffected by lactation tetany, whereas the disease is prevalent in the United Kingdom. In New Zealand, metabolic diseases are complex and the incidence is high, both of which are probably related to the practice of having the cows calve in late winter when feed is poor, the practice of depending entirely on pasture for feed, and the high proportion of Jerseys in the cattle population. Detailed knowledge of the nutrition and housing factors is essential before any reasonable scheme of prevention can be undertaken. For example, knowledge of the complex behavioral needs of the dairy cow is essential to provide adequate housing during the transition period. In North American dairy herds, the flow of cows through the transition period often necessitates many changes of pens, which are disruptive to the social organization of cow groups. Stocking rates that exceed stall and feed bunk capacity place even greater challenges on the dairy cow at this time. Current free-stall recommendations include providing 75 cm (30 in.) of bunk space for close-up and fresh cows to ensure that overcrowding does not occur, moving preparturient cows to a new pen at least 8 to 10 days ahead of the anticipated calving date or when calving is imminent, adding cows to groups on a minimum of a weekly basis (it takes up to 1.5 days for a new social order to be determined after the addition of a new animal), and keeping a clean and comfortable environment. The diagnosis and treatment of dairy cows with periparturient diseases requires a program suited to the particular herd. Particularly in large herds, there is a need for collaboration between the veterinarian, nutritionist, manager of the herd, and animal attendants. Specific procedures should be developed for each herd based on past experience with the problems of recently calved cows, the facilities, the skills of the workers, the priorities of management, and the flow patterns of the cows in the herd. Every effort must be made to prevent periparturient diseases in the cows. In general, diseases in the

early postpartum period originate in the feeding and management of the dry cow. Important principles include a protocol of grouping parturient cows according to the feeding program and handling facilities on the farm. Groups of cows can be screened for mastitis, visual evidence of illness, daily milk yield, body temperature, and urine pH, and they can be palpated for evidence of metritis. Individual cows that have been identified by a screening method must be examined individually to make a diagnosis and decide on a treatment protocol based on the particular diagnosis. Management and environmental factors can be manipulated to ease the transition into lactation. For example, the photoperiod, defined as the duration of light exposure an animal receives within a day, can be adjusted to produce clinically significant effects on periparturient health and subsequent lactational efficiency. Increasing the frequency of milking in the immediate postpartum period also produces persistent increases in milk yield and improvements in mammary health. In both techniques, evidence is emerging to support the concept that alteration of prolactin sensitivity is the mechanism underlying health and production responses. The reader is directed to publications related to production for more information on these and related topics. Occurrence and Incidence of Metabolic Diseases Knowledge of the etiologic and epidemiologic factors involved will help in understanding the occurrence and incidence of the various metabolic diseases. Largely because of variations in climate, the occurrence of metabolic disease varies from season to season and from year to year. In the same manner, variations in the types of disease occur. For example, in some seasons, most cases of parturient paresis will be tetanic; in others, most cases of ketosis will be complicated by hypocalcemia. Further, the incidence of metabolic disease and the incidence of the different syndromes will vary from region to region. Ketosis may be common in areas of low rainfall and on poor pasture. Lactation tetany may be common in colder areas and where natural shelter is poor. Recognition of these factors can make it possible to devise a means whereby the incidence of the diseases can be reduced. The metabolic diseases, because of high prevalence and high mortality rate, are of major importance in some countries, so much so that predictive systems are being set up. Rapid analysis of stored feed samples, pasture, and soil is commonly used in Europe and North America, but the interesting development has been the recognition of “production diseases” and the consequent development of metabolic profile tests, particularly in the United Kingdom and Europe.

Metabolic Diseases of Ruminants

VetBooks.ir

Record Keeping The use of reliable records to monitor the health and production of dairy cows during the transition period is essential to evaluate the efficacy of programs at the farm level. Transition cow management programs will assist in determining how well cows are prepared for milk production and good health in the coming lactation. Appropriate monitoring should focus on three areas: cows that die or are culled in early lactation, the productivity of the surviving cows in early lactation, and the rates of disease in the periparturient period. Cows that leave the herd in the first 60 days of lactation are usually culled because of disease or injury. Removal rates and their causes can be a critical monitor of the efficacy of transition cow management programs. Measuring productivity and health of cows in early lactation involves monitoring daily milk yields, first-test mature-equivalent 305-day projected milk, milk components at first Dairy Herd Improvement Association (DHIA) test day, milk-fat percentage, ratios of test-day components, somatic cell count at first DHIA test day, and peak milk (also called summit milk). DHIA records also allow comparison of the performance of each cow in early lactation to her performance in the prior lactation. Comparisons can be made of the changes in somatic cell count between the last test of the prior lactation and the first test of the current lactation and mature-equivalent 305-day difference from the prior lactation to the first test of the current lactation. Health and production records in dairy herds have traditionally emphasized reproductive events and administered treatments for specific diseases. The records should capture the information about the common diseases that occur in most dairy herds. The record system should be set up to do the following: • Monitor rates of well-defined disease events as a measure of the effectiveness of health and production programs and to aid in problem solving. • Determine the clinical efficacy of treatments by monitoring retreatment rates for specific diseases. • Maintain an individual cow history record for cow-side use to enhance treatment decisions. • Measure compliance and consistency of implementation of the health program being used. • Reconcile pharmaceutical purchases with treatment protocol entries and to meet regulatory requirements on the use of pharmaceuticals in food animals. • Determine the costs of certain disease rates over achievable targets. The costs of specific diseases are compelling to most dairy herd producers. Good records can generate an incidence rate of common diseases. These costs include the

immediate cost of treatment, the cost of the veterinarian’s and herdsman’s time. and the cost of milk withheld from the market. For the majority of diseases of recently calved cows, the cost per disease in the United States was estimated in 2001 at approximately US$320, with a range from $150 to $450. An adequate record system will allow producers and veterinarians to determine the differences between actual performance and benchmark performance and then determine the causes of the shortfall. The most important determinants of profitability on dairy farms are milk income and feed cost, and the difference between milk income and feed costs is the return-over-feed index (ROF). Many factors affect the ROF index. These include three-times-daily milking, component percentages in the herd milk test, milk-fat and protein percentages, use of a core lipopolysaccharide antigen mastitis vaccine, and use of monensin in the lactating-cow diet (if permitted). One of the most important factors associated with profitability is milk production. From 80% to 95% of the income on dairy farms is derived from milk sales. Thus it is critical that the producer, the veterinarian, and other advisors collaborate to plan an animal health and production program that will result in the optimum ROF.

METABOLIC PROFILE TESTING Methods are needed to monitor the nutritional and metabolic status of dairy herds. The most valuable methods will be those that are sensitive enough to detect change before clinical or economic consequences are manifested. A major challenge in the application of metabolic profile testing is dealing with extraneous sources of variation. Successful management of extraneous variation requires sampling strategies based on animal grouping and testing of multiple animals. Larger herds are more suitable for monitoring because they allow for better design sampling strategies and spread the costs of testing across more animals. Statistical process control methods offer a unique approach to interpretation that may increase the usefulness of metabolic profiles. The traditional approach to herd-based assessment of metabolic status (also called the Compton metabolic profile test) is based on the concept that the laboratory measurement of certain components of plasma or serum of 7 to 10 cows per subgroup will reflect the nutritional status of the subgroup, with or without the presence of clinical abnormalities.1 For example, a lowerthan-normal mean plasma glucose concentration in a group of dairy cows in early lactation may indicate an insufficient intake of energy, which may or may not be detectable clinically. On a theoretical basis, the ability of the laboratory to make an objective

1667

assessment of the input–output (nutrient– productivity) relationships is an attractive tool for the veterinarian engaged in providing a complete health management service to a herd. The test would theoretically be able to detect the qualitative and quantitative adequacy of the diet of cows expected to produce a certain quantity of milk or return to estrus within a desirable length of time following parturition. A reliable test for the early diagnosis of nutritional deficiency or metabolic disease would therefore be a major step forward in attempting to optimize livestock production and obtain maximum yields at minimum costs. There was considerable interest in metabolic profile testing following its earlier descriptions, which stimulated considerable field research. The results of the research have thus far indicated that the test may be useful only as an aid in the diagnosis of nutritional imbalance and production diseases. The results of metabolic profile testing are usually difficult to interpret without a careful conventional assessment of the nutritional status and reproductive performance of the herd, and it appears doubtful that such testing would reveal significant abnormalities that could not be detected using conventional clinical methods. Because of the cost of the test, the profile testing must be carefully planned with specific objectives. A regional diagnostic laboratory with automated analytical equipment should be available, and this is often a major limiting factor. The test should not be undertaken unless reference values for each laboratory measurement are available from the population within the area. The results from the groups within the herd are compared with local population means. Metabolic profiles have also been suggested as an aid in the selection of superior individuals. The prediction of whether an individual cow is metabolically prepared to undergo a stressful lactation at a high level of production would seem to be a useful undertaking. This could be particularly important under management conditions of heavy concentrate feeding, lead feeding, zero grazing, or even indoor housing. There are no wellestablished, low-cost, practical protocols for conducting such profile tests. Usefulness of Metabolic Profile Testing Metabolic profiles in dairy cows were used initially in the United Kingdom in the 1960s. Success was limited primarily by the unjustified expectation that all biochemical concentrations in the blood of cows would reflect nutritional intake and status at all times. However, the practical value was found in the approach as an aid to nutritional management. In the 1970s the approach was reassessed and revised, culminating in a program for farmers evaluating health and productivity using metabolic profile testing as an

1668

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

integral part of a health management program involving a multidisciplinary approach. The UK Dairy Herd Health and Productivity Service (DHHPS) provides the opportunity for veterinarians to lead a multidisciplinary team that can monitor health, fertility, and production and can plan, when necessary, corrective action. Effectively the approach has been to “ask the cows” what they think of their nutrition by following a set of guidelines on timing, cow selection, and the use of background information. Metabolic profiling and body-condition scoring found that at least a third of the cows sampled were mobilizing excessive fat during the transition from the dry period to early lactation. Improving both health and nutrition, before and after calving, would improve reproductive performance in many herds. The DHHPS method now utilizes a team approach involving farmer, veterinarian, and agricultural advisor. If useful information is to be obtained, the blood-testing aspect depends critically on following a set of firm criteria for selection of small groups of typical cows within each herd; the timing of testing in relation to concentrate feeds, feed changes, and stage of lactation; and the collection of other data about the cows, such as body weight and condition, productivity, and feeding. The successful approach has been to look, following specific times of nutritional change, at metabolite levels in strictly defined small representative groups of cows within each herd in conjunction with information on body condition and weight, milk performance, and feeding. Comparison with optimum values, the degree of variation from them, and comparisons between groups within herds have allowed information about nutritional constraints on productivity to be made available to farmers more quickly and more specifically than by other means. Biological and Statistical Basis for Herd Testing The interpretation of herd-based tests for metabolic diseases is different from interpreting laboratory tests for metabolites from individual cows. Test results from individual cows are interpreted by comparing the value to a normal reference range established by the laboratory that did the testing. Normal ranges are often derived by calculating a 95% confidence interval (or a similar statistic) of test results from 100 or more clinically normal animals. Herd test results for metabolic diseases can be interpreted as either the mean test result of the subgroup sampled or as the proportion of animals above or below a certain cut-point within the subgroup. There is a major philosophic difference and marked cost difference between the two approaches. At the moment, there does not appear to be a clear preference for either approach, and this area should be a focus of future investigation. An important difference

in the approaches is that the cost of determining the mean value of a subgroup is very low if the samples from the subgroup are pooled and then analyzed. The only advantage of analyzing individual samples instead of pooled samples is that individual samples provide an estimate of the proportion of abnormal values. Whether knowing the proportion is worth the marked increased in analytical cost remains to be determined. If a metabolite is associated with disease when it is above or below a biologic threshold (cut-point), then it should be evaluated as a proportional outcome. For example, hyperketonemia (subclinical ketosis) in dairy herds can be monitored by testing for β-hydroxybutyrate (BHB) or other ketone bodies in blood, plasma, serum, urine, or milk. Subclinical ketosis is a threshold disease, and cows are affected only when ketone concentrations are elevated. Plasma BHB concentrations above 1.0, 1.2, or 1.4 mmol/L (equivalent to 9.7, 11.7, or 14.4 mg/dL, respectively) are the most commonly used cut-points for detecting hyperketonemia (subclinical ketosis) in lactating dairy cattle. Early-lactation cows with plasma BHB concentrations above the selected cutpoint have a four- to eightfold greater risk of developing displaced abomasum, a threefold greater risk of developing clinical ketosis, decreased 305-day milk production, increased severity of mastitis, a 50% increase in anestrus at 60 days in milk, and a 50% decrease in pregnancy at first insemination. NEFA concentrations in plasma are an indicator of negative energy balance in prepartum cows. Elevated plasma NEFA concentration before calving (>0.4 mEq/L for cows between 2 and 14 days before the anticipated calving date) is associated with a 4-fold increased risk for displaced abomasum, a 2- to 3-fold increase in the risk of subclinical ketosis, and 1.5-fold increased risk of retained placenta after calving. It is also necessary to determine the alarm level for the proportion of animals above or below the described cut-point. The alarm level is determined from research results or clinical experience. The suggested alarmlevel proportion for plasma BHB concentration with a cut-point of 1.4 mmol/L is greater than 10%; the proportion for plasma NEFA concentration with a cut-point of 0.4 mmol/L is greater than 10%. Herd-based testing is useful only when a sufficient number of cows within the herd are tested, which gives reasonable confidence that the results truly represent the entire population of eligible cows in the herd. In the United States, the minimum sample size for herd-based tests with proportional outcomes has been estimated as 12 cows, based on 75% confidence intervals and a general detection cut-point of greater than 10%. Cows to be sampled need to be selected from the appropriate eligible or at-risk group. Obviously, measurement of a homogeneous group

requires a large herd size; to sample 12 cows consistently between day 4 and day 14 of lactation requires a milking herd of at least 428 cows, assuming equal monthly calving rates. The subgroup size recommendation of 12 cows in the United States contrasts with the subgroup size recommendation of 7 to 10 cows in the United Kingdom. The difference in recommendations has not been reconciled. The proper use of metabolic profiles depends on the timing of blood tests, the selection of cows to be included, and the collection and use of background information about the farm, feeding and feeding system, and physical state and performance of the cows. Variables in Dairy-Herd Metabolic Profile Testing There are five main areas of interest for metabolic profile testing: 1. Energy balance 2. Protein evaluation 3. Liver function 4. Macromineral evaluation 5. Urine evaluation In general terms, measurement of analytes in urine has been greatly underutilized in metabolic profile testing, and it is clear that current testing protocols are not economically optimized. Energy Balance Strategic use of metabolic testing to monitor transition dairy cows should focus on measuring plasma NEFA concentration in the last week prepartum and plasma/serum BHB and urine acetoacetate concentration in the first and second weeks postpartum. Nonesterified Fatty Acids Plasma NEFA concentration provides the most sensitive indicator of energy balance, particularly in the last 2 weeks of gestation. Plasma NEFA concentration is useful for monitoring the energy status of dry cows in the last month of gestation, when rapid changes in energy-balance status may not be detectable from changes in body-condition score. Plasma NEFA concentrations start to increase 3 days before parturition and remain elevated for the first 9 days of lactation7 (Fig. 17-2). High plasma concentrations of NEFAs indicate negative energy balance, which occurs in animals that are inappetent as a result of illness. The serum concentrations of NEFAs have been monitored in dairy cows as predictors of displaced abomasum. In cows with leftdisplaced abomasum (LDA), mean NEFA concentration began to diverge from the mean in cows without LDA 14 days before calving, whereas mean serum BHB concentrations did not diverge until the day of calving. Prepartum, only NEFA concentration was associated with risk of LDA. Between day 0 and 6 days after calving, cows

Metabolic Diseases of Ruminants

ketosis). In cows with serum BHB con­ centrations of 1.2 mmol/L or greater or 1.4 mmol/L or greater in the first week postpartum, the odds of LDA were three and four times greater, respectively, than in cows with BHB below the cut-points.7

0.9

VetBooks.ir

0.8

NEFA, mmol/L

0.7 0.6 0.5 0.4 0.3 0.2 0.1 0

A

–21 –18 –15 –12

–9

–6

–3

0

3

6

9

12

15

18

21

–21 –18 –15 –12

–9

–6 –3 0 3 6 Day relative to calving

9

12

15

18

21

1.2

BHBA, mmol/L

1.0 0.8 0.6 0.4 0.2

B

1669

0.0

Fig. 17-2  Least-squares means and 95% confidence interval (error bars) for plasma nonesterified fatty acid concentration and β-hydroxybutyrate concentration in 269 multiparous Holstein–Friesian cows from 21 days prepartum to 21 days postpartum. (Reproduced with permission from McCarthy MM, Mann S, Nydam DV, Overton TR, McArt JAA. J Dairy Sci 2015; 98:6284-6290.)

with serum NEFA concentration of 0.5 mEq/L or greater were 3.6 times more likely to develop LDA after calving. In another study, cows with plasma NEFA greater than 0.3 mEq/L between 3 and 35 days before calving were twice as likely to subsequently have LDA. Strategic use of metabolic tests to monitor energy balance in prepartum dairy cows should therefore focus on measurement of plasma/serum NEFA concentration. There are three major drawbacks with this approach: the high cost of testing (US$8/test), the need to centrifuge blood to harvest serum or plasma, and the lack of a cow-side test. Until all three issues are satisfactorily resolved, plasma/serum NEFA testing will remain a research tool with minimal practical application. β-Hydroxybutyrate Plasma/serum BHB concentrations are affected by energy and glucose balance and

are a less sensitive indicator of energy balance than plasma NEFA. High plasma BHB concentrations are associated with reduced milk production, increased incidence of clinical ketosis and LDA, and reduced fertility. The gold-standard test for hyperketonemia (subclinical ketosis) is plasma/serum BHB concentration, which is more stable after collection than plasma/serum acetone or acetoacetate concentrations. Prepartum BHB concentrations are relatively stable before parturition, but increase rapidly after parturition to peak at around 9 days in milk, after which time BHB concentration gradually declines (Fig. 17-2). Subclinical ketosis may start at serum concentrations above 1.0 mmol/L. The alarm level for the proportion of cows above the cut-point of 1.0, 1.2, or 1.4 mmol/L has not been validated, but it is suggested that no more than 10% of early-lactation cows should have hyperketonemia (subclinical

Glucose Plasma/serum glucose concentrations are usually lower in early lactation4 and during the winter months; in early lactation, there is a heavy demand for glucose, and during the winter the energy intake is likely to be lower than necessary to meet requirements. One major cause of variation in blood glucose may be the major fluctuations in daily feed intake. Investigations of feed intake of dairy cows on commercial farms have shown that concentrate dispensers are commonly incorrectly adjusted, and errors of more than 50% in feed intake are sometimes found. In situations of marginal energy imbalance, glucose concentrations may be unreliable as an index of the adequacy of energy intake. Several factors may cause short-term changes in glucose concentration. Blood glucose may be influenced by the chemical nature of the carbohydrate and physical form of the feed and the roughage content of the feed. In addition, elevation of plasma glucose concentration has been associated with excitement and low environmental temperature. There is some conflicting evidence about the relationship between the mean plasma glucose concentrations of a lactational group and insufficient energy intake and reproductive inefficiency. In some work, there is an expected relationship between low plasma glucose concentration and an increased incidence of ketosis. In others, the relationship is not clear; however, there was a more consistent relationship between the actual energy intake as a percentage of requirement and the plasma NEFA concentration, but this finding was not sufficiently reliable to be useful. The mean plasma glucose concentrations within 3 days before or after first service of cows that conceived on first service was higher than that of cows that returned, but the difference was only approaching significance at the 5% level, and it is doubtful whether this could be of practical value. Although plasma NEFA concentration is more sensitive than plasma glucose concentration as an indicator of energy status of the lactating cow, the excessive variability of this relationship during early lactation limits its usefulness. Plasma NEFA concentration begins to increase several weeks prepartum, peaks at parturition, and decreases gradually to normal concentrations after several weeks of lactation. Plasma glucose concentrations follow a similar pattern. The main disadvantage of using plasma glucose concentration as an index of metabolic balance is that glucose concentration is a tightly regulated variable, and marked metabolic imbalances need to be present before plasma glucose

1670

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

concentration is altered. However, this means that decreased plasma/serum glucose concentrations provide an unequivocal and specific indicator of negative energy balance. Protein Evaluation Currently, there is not a single biochemical factor that accurately reflects the protein status of dairy cattle. A number of indices have therefore been monitored, including milk urea nitrogen and plasma/serum urea nitrogen, creatinine, albumin, and total protein concentrations. Of these indices, milk urea nitrogen concentration in the bulk tank provides the best global picture of protein balance in a dairy herd. Urea Nitrogen Plasma urea nitrogen and milk urea nitrogen (MUN) concentration are useful indicators of protein status, particularly when the diet contains adequate energy. Increases in plasma urea nitrogen concentration and ammonia occur primarily as a result of inefficient nitrogen utilization. An excess of rumen degradable protein results in an increase in the concentration of rumen ammonia, which is absorbed through the rumen wall and transported to the liver, where it is converted to urea. The catabolism of body protein for gluconeogenesis can also result in the production of ammonia, which is also converted to urea in the liver. Plasma urea nitrogen concentration has therefore been the most commonly used blood constituent for monitoring protein status and intake. Values greater than 19 mg/dL suggest excessive protein intake in the diet, whereas values less than 10 mg/dL suggest inadequate protein intake in the diet. MUN concentration can be used as a management aid to improve dairy-herd nutrition and monitor the nutritional status of lactating dairy cows. Elevated MUN concentration indicates that excess protein has been fed to the dairy cow for a given level of production. Milk samples should be submitted to an accredited diagnostic laboratory for MUN analysis. The Azotest Strip, an on-farm dipstick test, lacks accuracy and is not recommended. The MUN target concentrations for lactating dairy cows fed according to National Research Council recommendations have been evaluated. The target MUN concentrations are 8.5 to 11.5 mg/dL for most dairy herds compared with the previous target concentrations of 12 to 16 mg/dL. MUN, together with percentage milk protein, is being used increasingly as an indicator of the dietary protein–energy balance. The time of sampling can have a significant effect on MUN concentrations; the highest concentration was found to occur in the morning, and the diurnal pattern was not influenced by intrinsic factors such as parity, days postpartum or daily milk yield. MUN concentration was significantly increased after refrigeration for 1 week.

Several reviews of the literature have examined the effect of protein nutrition on reproduction in dairy cows. The reported effect of high nitrogen intake on fertility is inconsistent. Experimentally, the ingestion of a high level of degradable protein commencing 10 days before insemination in lactating dairy cows had no effect on the reproductive performance of the lactating high-yielding dairy cow. The relationship between MUN concentration and the fertility of dairy cows from 250 herds in the United Kingdom found no relationship between bulk-tank MUN concentration and fertility, or between changes in bulk-tank MUN concentrations and fertility. A meta-analysis of the literature evaluated the associations between dietary requirements for protein for dairy cattle, the metabolism of protein in cattle, factors influencing the degradability of protein in ruminant feeds, and factors influencing MUN concentrations. There are good correlations between dietary protein intake and rumen ammonia, blood urea, and milk urea concentrations. Ryegrass clover pastures provide feed in many of the temperate dairy regions of the world, and for much of the year pasture crude protein may exceed 30%, of which a high proportion is rapidly degradable. High dietary protein intakes may have a negative effect on reproductive performance in lactating dairy cows, but the role of milk urea as a predictor of fertility needs further definition given the high conception rates in many Australasian dairy herds. High intakes of dietary protein may induce adaptations in urea metabolism, and the negative relationship identified between high intakes of dietary protein and fertility for northern hemisphere dairy herds may not necessarily apply in Australasian dairy herds. Because of the potential for cows to adapt to high-protein diets, the use of a single MUN determination on a herd will have limited value as an indicator of nutritional status and little value as a predictor of fertility. The differing observations between various production systems indicate the need for careful consideration in applying recommendations for dietary protein management based on milk urea concentrations. MUN determinations may, however, have value, particularly when used in conjunction with other herd and nutritional data to assess the protein nutrition of dairy herds. It is unlikely that single or even serial determinations of MUN concentration in single cows or bulk-tank milk will have a high predictive value for determining the risk of conception in the cow or herd. Albumin Plasma/serum albumin concentration is related to the protein status of the animal and whether an acute-phase reaction has been induced. Lactation stage has a substantial effect on serum albumin concentration. Animals should be grouped into dry cows,

early lactation (1 to 10 weeks), and later lactation. Minimal values for dry-cow means are from 2.9 to 3.1 g/dL, from 2.7 to 2.9 g/dL for recently calved cows, and from 3.0 to 3.2 g/dL for cows in later lactation. Liver Function and Injury The presence of liver injury can be evaluated by measuring plasma/serum aspartate amino-transferase (AST), sorbitol dehydrogenase (SDH), alkaline phosphatase, and gamma-glutamyl-transferase (GGT) activities. Of these enzyme activities, the plasma/ serum AST activity is the most clinically useful, with plasma AST greater than 162 U/L being indicative of hepatic lipidosis. Because increased AST activity also reflects damaged skeletal muscle, AST activity is not a specific test for liver injury in cattle at calving or for a few days after calving because of the potential for parturitionrelated muscle damage. Liver function can be assessed by measuring the plasma/serum total bilirubin, cholesterol, and albumin concentrations. Serum bile acid concentration is not a useful index of liver function in cattle. Calculation of the ratio of plasma NEFA to cholesterol on a molar basis appears to provide a clinically useful index to evaluate the degree of hepatic lipidosis and the liver’s ability to export mobilized peripheral fat reserves, and has been able to predict the incidence of postpartum disease. The major drawback with measuring the NEFA:cholesterol concentration is the cost, in that each analysis costs approximately US$8/test, for a total test cost of US$16. Plasma/serum albumin concentrations are decreased for the first month after calving as a result of plasma volume expansion to accommodate the nutrient flow for milk production, loss into the uterine lumen associated with uterine involution, catabolism of body protein resulting from negative energy balance, and decreased hepatic function as a result of hepatic lipidosis. The major clinical utility of plasma/serum albumin concentration is therefore in the first 4 weeks of lactation. Macromineral Evaluation Abnormalities of the blood levels of the four macrominerals, calcium, phosphorus, magnesium, and potassium, in the cow during the transition period are involved in subclinical hypocalcemia, clinical milk fever, hypomagnesemia, and acute hypokalemia. Calcium Serum calcium concentrations are tightly regulated and are not sensitive indicators of input–output balance. Measurement of plasma/serum calcium concentrations during the first 24 hours after calving, particularly in multiparous dairy cows, can provide useful insight into the effectiveness of control programs for periparturient hypocalcemia.

Metabolic Diseases of Ruminants

VetBooks.ir

Phosphorus Serum inorganic phosphate concentrations tend to fall following long-term insufficient dietary intake. Magnesium Serum magnesium concentrations are usually low during the winter months, and subclinical hypomagnesemia exists in many herds, especially pregnant beef cattle. This can be converted into clinical hypomagnesemia with a sudden deprivation of feed or a sudden fall in environmental temperature. Supplementation of the diet with magnesium salts is protective. Sodium Low serum sodium concentrations occur in early lactation in cows grazing on summer pastures without supplementation with salt. Levels down to 135 mmol/L may be associated with depraved appetite and polydipsia and polyuria. Potassium Serum potassium concentrations have been difficult to interpret because the levels of the electrolyte in serum are not necessarily indicative of potassium deficiency. The normal serum potassium concentration is much more variable than that of sodium, and its average concentration in roughages of all kinds is nearly always in excess of requirements; any abnormalities are usually in the direction of excess. Hematology Hematocrit (Packed Cell Volume) The hematocrit can be used as a general reflection of health. In most dairy herds, a low hematocrit may be a reflection of suboptimal energy and protein nutrition. Mean values of packed cell volume (PCV), hemoglobin, and serum iron are consistently higher in nonlactating cows than in lactating cows. Parasitism causing blood loss will result in a low hematocrit. The hematocrit varies with lactation stage, being highest in dry cows and lowest at peak (summit) milk. Cows should be grouped by lactation stage when evaluating hematocrit. Urine Evaluation Urine samples are easier to obtain than blood samples, although stimulation of the perineal area to induce urination is usually only 75% to 90% successful. Higher success rates in obtaining a urine sample are obtained in cattle that have been sitting down and that are encouraged to stand. Urine appears to be the optimal fluid to monitor acid–base and calcium status in dairy cattle. The most accurate insight into acid–base homeostasis in healthy cattle is obtained by measuring urine net acid excretion (NAE) or net base excretion (NBE) (see Chapter 5). However, when urine pH is between 6.3 and 7.6, urine pH measured by

urine dipsticks or pH papers provides an inexpensive and clinically useful insight into acid–base homeostasis in cattle.5 This is because the change in urine pH over this pH range accurately reflects the change in NAE or NBE. Optimum target values for urine pH to decrease the incidence of milk fever in dairy herds have not been identified, and recommendations for optimal urine pH values vary widely.5 Feeding rations with low dietary cationanion difference (DCAD) to dairy cows for at least 2 weeks before calving decreases the incidence of periparturient hypocalcemia. The most likely reason for this effect is that ingestion of a low-DCAD diet increases calcium (Ca) flux, which in nonlactating cows is most readily detected as an increase in urinary calcium excretion. Low urine pH decreases calcium uptake from the tubular lumen into the renal epithelial cell and therefore decreases calcium absorption in the distal convoluted tubule and connecting tubule, thereby directly resulting in hypercalciuria. It remains to be determined whether laboratory measurement of urinary calcium concentration is more accurate and costeffective than cow-side measurement of urine pH or laboratory determination of urinary strong ion difference and NBE when evaluating the effectiveness of milk-fever control programs. Timing of Blood Tests In Relation to Feed Changes Because changes in the diet of ruminants cause changes in the character of rumen activity, blood samples for metabolic profile testing should not be done until 2 weeks after a major dietary change. Minor changes such as an increase in the quantity of an existing component or in access to the same ration do not require a wait of more than 7 to 10 days. Changes in forage type, such as turnout to pasture, housing, or the introduction of silage, require the full 2 weeks. The same applies for introduction of concentrates or of a new type of concentrate. In Relation to Feeding There can be changes in biochemical values in blood associated with feeding. These are most marked in cows receiving their entire concentrate ration at milking time. In such cases, 2 hours should be allowed to elapse after milking before blood sampling. In circumstances where the major part of the concentrate input is mixed with the forages and is available for most of each 24 hours, the timing of tests in relation to feeding is less critical. If lower-yielding midlactation cows are included (see later discussion), their results can be used as a check to see if there is an effect of feeding on the biochemical values in the blood samples. Cows should not be separated at milking time and confined for hours without access to food

1671

while waiting for blood sampling because this can also affect the results. The available recommendations regarding the timing of blood collection relative to feeding are somewhat confusing. A common recommendation is that blood samples should be obtained 5 hours after feeding if animals are fed a fresh total mixed ration once daily. This recommendation does not seem logical in that plasma BHB concentration is increased at this time in cattle fed rations based on corn (maize) silage because of the metabolism of absorbed butyrate by ruminal epithelial cells. In most nutritional studies, blood samples are collected in a preprandial state because this is most consistent, but this will give the highest plasma NEFA concentrations over a 24-hour period. On this basis, sampling should be done in the morning immediately before or at the time of feeding of a fresh total mixed ration. In Relation to Calving Pattern and Seasonal Feeding Changes The cow in early lactation is the most important because what happens to her in the first few weeks after calving has the major influence on her subsequent productivity, including her future fertility efficiency. Therefore blood sampling for metabolic profiles should be carried out at the beginning of each new calving season, with the first cows checked so that the majority can benefit from the information derived. Of equal importance is the need to test as soon as possible after the introduction of a new ration, so that evaluation of the cows’ biochemistry can be made available as quickly as possible to determine what the cows, the end users, think of the ration. Therefore planning of metabolic profile tests needs to be done in advance and should take into account both expected calving pattern and feed changes. Without planning along these lines, time may be lost, and productivity with it. Selection of Cows Picking appropriate cows for blood sampling is very important. This is because some of the metabolites looked at, particularly those relating to energy balance, can quickly return to the optimum range as cows adapt themselves, including their productivity, to a nutritional constraint. It is possible for cows to experience a significant energy deficit in the first 2 to 3 weeks of lactation because of intake problems, lose excessive body condition, perhaps have their milk yield modified, and have their subsequent fertility efficiency suppressed, but yet still arrive at 4 weeks calved with all biochemical measurements within the optimum ranges. If blood is sampled at 4 weeks after calving or longer, a producer could see thin, underproducing cows with poor fertility but with nothing abnormal about their biochemistry. Thus the farmer would be entitled to feel the

1672

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

metabolic profile test was of no value. However, if those cows had been blood sampled at 14 days calved instead of 28 days, the blood results would have been quite different and would have identified the nutritional constraint on productivity. The guidelines for metabolic profiling of dairy cattle recommend sampling from the following groups: • Dry period (D): between 7 and 10 days before anticipated date of calving • Early lactation (EL): between 3 and 14 days of lactation • Midlactation (ML): between 50 and 120 days of lactation. Individual variations in biochemical values are such that single cows should not be tested. Groups of no less than five should be sampled. They should not be picked at random, but rather should be typical, average cows of their stage of lactation. Cows with extremes of performance—either very high or very low—should not be selected. Cows with problems should also not be included because the type of analysis carried out is not designed to clarify individual problems. It is important to make all this clear to farmers in advance because they cannot be expected to appreciate the limitations of the analyses made. Experience in the Dairy Herd Health and Productivity Service in the United Kingdom suggests that selecting cows for metabolic profiles may be best done by the veterinarian in advance of the test after looking at the calving and production records. If there is a specific concern of poor conception rate, for example, farmers may expect only cows that failed to conceive to be sampled. This tactic hardly ever delivers helpful information because any nutritional constraints have by then been compensated for, and blood biochemical values are usually within optimum ranges. The best approach may be to include such cows as the midlactation group. Dry-Cow Group Because the dry period is so important to the success of the following lactation, blood sampling to make sure nutrition is adequate is essential. However, the nature of the measurements that can be made means that primarily cows in the last 7 to 10 days of gestation should be sampled. Blood sampling a group of dry cows with 1 month or longer to go to calving at the same time can sometimes provide a useful within-herd comparison with respect to energy balance. Such sampling may also identify the presence of dietary protein inadequacy—specifically, rumen degradable—in the early part of the dry period. Early-Lactation Group The definition used for the early-lactation group is most critical for the reasons given in the previous paragraph. Since the original

Compton metabolic profile, in which a highyielding cow was used as the definition, the importance of this group has become increasingly apparent. The definition also has had to be changed to take into account changes in farm practice. The way cows are fed now—total mixed rations, increased outof-parlor concentrate feeding—has reduced the time after calving by when they can adapt themselves to an unsatisfactory diet. To be sure of detecting the presence of an energy constraint in particular, blood sampling should be carried out between 3 and 14 days calved—at less than 3 days, the metabolic impact of hypercortisolemia associated with parturition is still present; at more than 14 days, some cows will be thin, unproductive, and subfertile but may have compensated for their nutrition and thus have normal blood metabolite values. Midlactation Group Some cows that have past the period of peak yield and so past the greatest period of potential nutritional stress should always be included. They should be between 50 and 120 days calved so that they are still relatively high yielding. This group provides a within-herd comparison with the earlylactation cows. Without this it is very difficult to distinguish between problems caused by constraints on intake of food or protein and energy content; to identify changes in biochemical values caused by mistiming of tests in relation to feeding or by oddities in the diet, such as silage with a high butyric acid content; and to make judgments on concentrate/forage usage within the herd. In the DHHPS program, a majority of farms do metabolic profiles three to four times a year at critical times as a check, or “ask-the-cows-what-they-think” exercise. Thus metabolic profiles are used as part of a proactive preventive health and productivity program. Some of the larger farms may do more than 10 tests a year to cover feed changes and to check on the success of any corrective action. In the DHHPS program, a standard DHHPS metabolic profile includes analysis of plasma for NEFA, BHB, glucose, urea nitrogen (urea N), albumin, globulin, magnesium, and inorganic phosphate. Analyses for copper and glutathione peroxidase (GSHPx) are done on approximately one-third of samples received and thyroxine T4 on even fewer. Biochemical analysis is performed using two biochemical auto-analyzers, with standard internal controls. It also employs an independent, external quality control system. Derivation of optimum metabolite values are summarized in Table 17-2. They are BHB less than 0.6 mmol/L in dry cows and less than 1.0 mmol/L in cows in milk; glucose greater than 3.0 mmol/L; NEFA less than 0.5 mmol/L in dry cows and less than 0.7 mmol/L in cows

Table 17-2  Metabolic profile parameters in cattle—optimum values Parameter BHB   Milkers   Dry cows Plasma glucose NEFA   Milkers   Dry cows Urea nitrogen Albumin Globulin Magnesium Phosphate (inorganic) Copper Thyroxine T4 (iodine) GSHPx (selenium)

SI units Below 1.0 mmol/L Below 0.6 mmol/L Over 3.0 mmol/L Below 0.7 mmol/L Below 0.4 mmol/L 1.7–5.0 mmol/L Over 30 g/L Under 50 g/L 0.8–1.3 mmol/L 1.4–2.5 mmol/L 9.4–19.0 µmol/L Over 20 nmol/L Over 50 units/g Hb

in milk; urea N greater than 1.7 mmol/L; albumin greater than 30 g/L; globulin less than 50 g/L; magnesium greater than 0.7 mmol/L; phosphate greater than 1.3 mmol/L; copper greater than 9.2 µmol/L; glutathione peroxidase (GSHPx) greater than 50U/g HB; and thyroxine T4 greater than 20 nmol/L. Energy.  The data in Table 17-3 use only the cows fitting precisely the definitions of D, EL, and ML. The table shows that, overall, an average of 30% EL cows had metabolite results reflecting satisfactory energy status, as did 61% of ML cows and 43% of D cows. In both EL and ML groups, glucose is the metabolite most commonly outside its optimum range, followed by BHB and NEFA. The percentage of NEFA values above optimum is low in ML cows. The most common finding is high BHB and low glucose in the same cow. In tests showing most cows in an EL group having these results, there is usually one or two with high NEFAs as well. Some EL cows show only low glucose or only high NEFA. Where low glucose only predominates in EL cows, ML cows often show the same picture. Protein.  The plasma urea concentration results in Table 17-3 show that the EL stage is more vulnerable to low values than later in lactation, even though the cows would have been on the same diets in virtually every case. In fact, an even greater average percentage of the blood of 1361 cows sampled between 0 and 9 days after calving over the 5 years showed low urea concentration. The proportion of low-ureaconcentration results in D cows is high (Table 17-3). In addition to the category shown of 10 days or less before calving, 4335 cows were sampled at more than 10 days

Metabolic Diseases of Ruminants

1673

VetBooks.ir

Table 17-3  Annual (April–March) percentages outside optimum ranges of metabolite results in blood plasma in adult dairy cows8 EARLY LACTATION (EL) (10–20 DAYS CALVED)

MID LACTATION (ML) (50–120 DAYS CALVED)

1999 /00

2000 /01

2001 /02

2002 /03

2003 /04

1999 /00

2000 /01

2001 /02

2002 /03

β-hydroxybutyrate (BHB)

19.5

16.6

22.3

22.9

17.5

11.2

10.5

14.3

Glucose Non-esterified fatty acid (NEFA) One or more energy metabolite per cow Urea-nitrogen (UreaN) Number of cows

46.0 19.1 65

48.7 22.2 70

43.1 24.9 67

49.0 27.4 72

59.3 28.0 78

21.8 0.6 34

25.9 2.1 39

14.5 1.8 32

0.8 1295

17.3 1421

18.3 1248

16.7 1285

16.7 1530

4.4 914

6.4 1066

6.0 849

prepartum, and 22% of them had low plasma urea concentrations. Results outside the optimum ranges for albumin (0.6%), magnesium (2.5%), phosphate (1.0%), copper (10%), and GSHPx (3%) are relatively uncommon. Thyroxine T4 analysis was carried out in 836 samples on specific request, and only 3% were below optimum. Background Information So that full value can be obtained by the farmer from the metabolic-profile approach, information about the cows and the farm should accompany the blood samples to the laboratory. This should include cow identification; last calving date for milkers/expected for dry cows; body weight (calculation from heart-girth measurement with a weighband pulled to a constant 5-kg tension is the best because it is not affected by gutfill and usually most practical because no mechanical weighing device/crush is required); bodycondition score by a palpation method; current daily milk yield; expected current daily milk yield; lactation number; daily supplementary feed intakes; daily estimated forage intakes; analytical description of feeds; and current herd milk solids percentages. It is useful to have information on herd size, breed, feeding systems, and health and fertility. A note of what concerns the farmer has, if any, should also be made. Interpretation of Results at the Farm Circumstances where the diagnosis of a nutritional constraint from blood samples is clearly correct, but the cause(s) are unclear from a distance and could be many, are common. Therefore it is very important that a final interpretation of what is not working and what are the best and most economic solutions ought to be made at the farm with the information from the laboratory in hand. Farm advisory visits should be made

DRY (D) (7–10 DAYS PREPARTUM)

2003 /04

1999 /00

2000 /01

2001 /02

2002 /03

2003 /04

10.6

9.6

34.5

24.7

38.5

28.5

22.7

22.5 2.7 40

25.4 3.1 44

23.9 10.8 59

27.3 15.0 57

21.7 14.2 63

27.3 13.0 46

33.8 14.8 63

5.3 1179

5.6 1494

18.4 1160

20.4 1379

20.2 1253

20.8 1358

22.3 1543

as soon as the results are available and discussions made, including farm staff and any other advisors involved. Experience in the DHHPS suggests that such a team approach produces a more balanced strategy and is more beneficial than each party working in isolation. Written Advice Any advice given should be recorded concisely in writing and copies given to all participants on the farm. This ensures that the agreed path is followed, creates a record, and ensures that the fee is for something tangible. Milk Production, Activity Meters, and Rumination Monitors The application of real-time monitoring of dairy cattle has great potential to provide low-cost and immediate insight into health and production. Of all potential indices, daily milk production (relative to previous production during the lactation or the previous lactation, or to peers) appears to provide the most sensitive and specific measure of dry matter intake and health. Milk production can be measured noninvasively and at very low cost using automated procedures at milking time. As diagnostic algorithms are refined, it is likely that monitoring of daily milk production will provide the most practical and low-cost method for evaluating health and production and for early disease detection. Activity meters can detect standing and lying periods in cattle and from that information can infer time spent feeding in cows housed in free stalls or tie stalls. Rumination monitors detect the time spent ruminating each day, which is strongly associated with dry matter intake and health. Body-Condition Score Managing body reserves is critical for successful cow management and requires an

accurate assessment of the cow’s “condition.” Body-condition scoring is an important aspect of metabolic diseases of farm animals. Body weight alone is not a valid indicator of body reserves because cows of a specific weight may be tall and thin or short and fat. The energy stores may vary by as much as 40% in cows of similar body weight, which emphasizes the futility and inaccuracy of relying on body weight alone as an index of cow condition. In addition, because tissue mobilization in early lactation occurs as feed intake is increasing, decreases in body-tissue weight can be masked by enhanced fill of the gastrointestinal tract, so that body-weight changes do not reflect changes in adipose tissue and lean-tissue weight. There is a strong positive relationship (R2 = 0.86) between BCS and the proportion of physically dissected fat in Friesian cows. Therefore the visual or tactile (palpation) appraisal of the cow’s BCS provides a good assessment of body-fat reserves, ignoring—or minimizing the effect of—frame size and intestinal contents. Most animal and dairy scientists acknowledge the successful manipulation of BCS as an important management factor that influences or has a relationship to animal health, milk production, and reproduction in the modern dairy cow. For example, cows that lost 0.5 to 1.0 point in BCS between parturition and first service achieved a pregnancyto-first-service rate of 53%, whereas those losing more than 1.0 point achieved a rate of 17%. In a seasonal pasture-based system for Holstein–Friesian cows, it is necessary to maintain the BCS at 2.75 or greater during the breeding season. BCS is important in achieving good reproductive performance. Loss of body condition between calving and first service should be restricted to 0.5 BCS to avoid a detrimental effect on reproductive performance. BCS is a subjective method of assessing the amount of metabolizable energy stored

1674

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

1

2

3

4

5

6

7

8

Spinous to Transverse Overhanging shelf Tuber coxae (hooks) Between pins Between Tailhead to pins SCORE Spinous processes SP (anatomy varies) Transverse processes processes (care-rumen fill) & Tuber ischii (pins) and hooks the hooks (anatomy varies) SEVERE UNDERCONDITIONING (emaciated)

1.00

individual processes distinct, giving a saw-tooth appearance

deep depression

very prominent >1/2 length visible

definate shelf, gaunt, tucked

extremely sharp, no tissue cover

prominent shelf

prominent

1.25

severely bones very prominent severe depression, depressed with deep “V” shaped devoid of cavity under tail flesh

1.50 1/2 length of process visible

1.75

FRAME OBVIOUS

2.00

individual processes evident

obvious depression between 1/2 to1/3 of process visible

2.25 2.50

bones prominent “U” shaped cavity formed under tail

very sunken

thin flesh covering

sharp, prominent ridge

definite depression

1/3 –1/4 visible moderate shelf

first evidence of fat

2.75 FRAME & COVERING WELL BALANCED

smooth concave curve

3.00

smooth ridge, the SP’s not evident

slight shelf

smooth

appears smooth, TP’s just discernable

3.25 3.50

100 g

1677

Ca/d), the active absorption of Ca from the digestive tract and mobilization from bone are homeostatically depressed and become quiescent. As a consequence, at calving when sudden changes of the Ca balance occur, the cow is unable to rapidly return to bone Ca stores or intestinal Ca absorption mechanisms and is susceptible to severe hypocalcemia until these mechanisms can be activated, which may take several days. Feeding prepartum diets with a Ca concentration low enough to induce a negative Ca balance already before calving prevents milk fever by activating Ca transport mechanisms in the intestine and bone before parturition, thus allowing the animal to adapt more rapidly to the lactational drain of Ca. The challenge of this approach is to formulate a balanced dry-cow ration with a sufficiently low Ca concentration providing less than 20 g/d of absorbable Ca (see also the discussion under “Control” in this chapter). Supplementing dietary Ca immediately before and around parturition may also lower the incidence of milk fever by providing additional dietary Ca at the moment intestinal Ca absorption is upregulated (see also the discussion under “Control” in this chapter). Dietary Potassium.  The dietary potassium content of the ration fed in late gestation is a major contributing factor to the risk of periparturient hypocalcemia. Positively charged electrolytes (cations) contained in the diet and absorbed from the digestive tract tend to alkalinize the organism, whereas electrolytes with a negative charge (anions) are acidifying.6 Studies of the 1970s furthermore demonstrated that alkalization of the organism by increasing the amount of dietary cations increased the incidence of milk fever, whereas acidification by increasing the dietary anion content reduced the milk-fever incidence, which is the basis of the so-called dietary cation–anion difference (DCAD) concept (see also the discussion under “Control” in this chapter). With potassium being the quantitatively most important cation in a standard ruminant diet, it follows that high potassium concentrations (>2% of the ingested dry matter) in the ration fed to cows in their last weeks of gestation can considerably increase the incidence of periparturient hypocalcemia. Dietary Magnesium.  Magnesium deficiency during late gestation is a major risk factor for periparturient hypocalcemia, and hypomagnesemia is considered to be the most common cause of milk fever occurring in midlactating dairy cows.3 Because magnesium is required for the release PTH from the parathyroid glands and furthermore influences the tissue sensitivity to PTH, the efficacy of PTH for the correction of hypocalcemia greatly depends on an adequate supply of magnesium.

1678

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Dietary Phosphorus.  Prepartum diets high in phosphorus (P; > 80 g P/d) greatly increase the incidence of milk fever and the severity of hypocalcemia. High dietary levels of P increase the serum level of P, which is inhibitory to the renal enzymes that catalyze the conversion of vitamin D to its active form 1,25-(OH)2D. Decreased amounts of 1,25(OH)2D will not only reduce the intestinal absorption of P, but also of Ca, and thereby predispose to periparturient hypocalcemia.7 Dietary Cation–Anion Difference.  Studies in the 1960s and 1970s showed that experimentally increasing the dietary cation concentration or decreasing the dietary anion concentration had an alkalinizing effect on the organisms, whereas feeding diets with a higher anion or lower cation concentration resulted in acidification. It was furthermore demonstrated that alkalization achieved through feeding high-cation/low-anion diets increased the incidence of milk fever, whereas acidification by increasing the ration of anions to cations caused the milk-fever incidence to decline.6 Accordingly the basis of the DCAD concept is to modulate the ratio of cations to anions in the diet to reduce the risk of milk fever in cattle. The cation– anion difference of a diet is commonly given in milliequivalents (mEq) per kilogram of feed dry matter (DM; mEq/kg DM) or sometimes in mEq/100 g DM. Although several different equations have been proposed for the calculation of the DCAD, the most commonly used only considers the four quantitatively most important dietary ions: sodium (Na+), potassium (K+), chloride (Cl–), and sulfur (S2–). Other electrolytes, such as Ca, P, and magnesium, also affect the actual DCAD and thereby the effect on the acid–base status of the animal consuming the diet, and these compounds are included in some of the equations proposed in the literature to calculate the DCAD. However, because these minerals are present in relatively low amounts in the ruminant diet, their effect is considered to be minor. They are therefore commonly disregarded for practicability reasons. The DCAD exerts a strong linear effect on the incidence of milk fever and is more important than the level of dietary Ca as a risk factor for milk fever. Prepartum diets high in cations such as potassium are associated with an increased incidence of milk fever, whereas diets high in anions, especially chloride and sulfur, result in a decrease in the incidence of the disease. Most forages, such as legumes and grasses, are high in potassium and are alkaline. The addition of anions, specifically chloride and sulfur, to the diet of dairy cows before parturition can effectively reduce the incidence of milk fever (see also the discussion under “Control” in this chapter). Systemic acidification induced by anionic supplementation affects the function of PTH hormone, the major effect being an increased

tissue response to PTH, which results in increased retention of Ca and enhanced mobilization of Ca from bone. A meta-analysis of 75 feeding trials designed to study the nutritional risk factors for milk fever in dairy cattle found that the prepartum dietary concentrations of S and dietary anion–cation balance ([Na + K]—[Cl + S]) were the two nutritional factors most strongly correlated to the incidence of milk fever. Dietary S acts as a strong anion and reduces the risk of milk fever, and increasing the dietary S concentrations lowers the odds ratio of developing milk fever.

ECONOMIC IMPORTANCE

Although economic losses from milk fever have decreased considerably since the introduction of intravenous treatment with Ca salts many years ago, the disease incidences reported in recent years remain similar to values reported decades ago.2,5 The most obvious costs are associated with drugs, veterinary intervention, and losses resulting from complications in clinical cases. Costs associated with subclinical hypocalcemia are, however, considered to be far more important. Incidence rates between 3.5% and 7% for clinical disease and over 30% for subclinical hypocalcemia have been reported, and hypocalcemia is considered to be a socalled “gateway disease” that predisposes to a number of common fresh-cow disorders, such as dystocia, uterine prolapse, retained fetal membranes, mastitis, ketosis, abomasal displacement, ketosis, and immune suppression.5 The costs per clinical case of milk fever have been estimated at US$300, whereas subclinical cases may cost around US$125 per case, based on estimates accounting for reduced milk production and increased risk of developing periparturient disorders such as ketosis or abomasal displacement.8 The literature on the effects of clinical and subclinical hypocalcemia is difficult to interpret because of the complex relationships between milk production, parity of lactation, breed of cattle, epidemiologic methods used, and management systems used, in addition to the reproducibility of the clinical observations and the accuracy of the recording systems used. In general, there is insufficient information available to document the consequences of milk fever and subclinical hypocalcemia. A summary of several consequences that have been examined follows here.

loss. The literature reports incidence rates for the downer-cow syndrome ranging from 3.8% to 28.2% of milk-fever cows, with a case fatality rate of 20% to 67%.9 Dystocia and Reproductive Disease.  Hypocalcemia at the time of parturition can result in uterine inertia, which may cause dystocia and uterine prolapse. In general, there is an increased risk of dystocia associated with milk fever, whether the farmer or the veterinarian attends to the dystocia.5 Retained Placenta.  Several studies have found an increased risk of retained placenta following milk fever. Metritis.  A few studies have found an indirect relationship between milk fever and subsequent metritis. Milk Production.  There is no reliable evidence that the occurrence of milk fever or subclinical hypocalcemia in cows that recover following treatment affects milk production in the subsequent lactation. Some studies have found a limited effect, no effect, or even positive effect of milk fever on milk production. Mastitis.  Hypocalcemia not only impairs immune function but furthermore may weaken the tone of the teat sphincter, which has been proposed to facilitate intramammary infection, particularly in recumbent cows that are not milked or milked less frequently.3 An odds ratio of 8.1 for mastitis has been estimated, for coliform mastitis the odds ratio is estimated at 9.0, and for acute clinical mastitis a relative risk of 1.5 following milk fever has been found. Displacement of Abomasum.  Odds ratios ranging from 2.3 to 3.4 for left-side displacement of the abomasum occurring in dairy cows with hypocalcemia at parturition have been estimated. Ketosis.  Studies on the occurrence of ketosis following milk fever have found relative risks or odds ratios ranging from 1.3 to 8.9; using all the confidence intervals, the relative risks/ odds ratios range from 1.1 to 15.3.

Milk Fever Relapses.  Milk fever cases that need repeat treatment because of relapses increase the costs.

Body Weight.  A temporary drop in body weight occurs in cows with milk fever, but there is no long-term effect. In cows with subclinical hypocalcemia in early lactation, there may be some weight loss compared with cows with normal levels of calcium.

Downer-Cow Complications.  The downercow syndrome associated with those milk fever cases that fail to respond to intravenous (IV) Ca treatment and remain recumbent for days before subsequently standing, those that die, or those that require destruction represents an important cause of economic

Culling.  There may be an increased probability of culling cows that have had milk fever because of the complications or direct or indirect consequences associated with the disease. There is some evidence of culling cows in early lactation because of milk fever, but not in late lactation.

Metabolic Diseases of Ruminants

PATHOGENESIS

VetBooks.ir

Hypocalcemia Calcium has several functions relevant for the pathophysiology of periparturient hypocalcemia, which include the following: • Cell membrane stability: Calcium bound to cell membranes contributes to the maintenance of adequate membrane stability. In excitable cells the decreased availability of ionized Ca results in higher cell membrane permeability, thereby altering the resting membrane potential and making nerve cells more excitable. • Muscle contractility: Calcium is required to clear the binding site for myosin on the actin molecule inside the muscle fibers. The cross-bridging between actin and myosin is the basis for the contraction of muscle fibers. Decreased availability of Ca can therefore affect muscle contractility. • Release of acetylcholine: Calcium is required for the neuronal release of the neurotransmitter acetylcholine into the synaptic cleft of the neuromuscular junctions. Calcium depletion can thus hamper the signal transmission at the level of the neuromuscular endplate. Whereas decreased membrane stability and ensuing increased excitability are the probable underlying cause of hypocalcemic tetany of monogastric species and the muscle twitching observed in the early stages of milk fever in cattle, disturbed muscle fiber contractility and neurotransmitter release are considered the basis of the flaccid paresis observed in advanced stages of milk fever in ruminants. The plasma Ca concentration is normally maintained between 2.1 and 2.6 mmol/L (8.4 to 10.4 mg/dL). Almost all multiparous dairy cows will experience at least transient and subclinical hypocalcemia, less than 1.8 mmol/L (7.5 mg/dL), within 24 hours after calving. In some cows, hypocalcemia is more pronounced, causing neuromuscular dysfunction resulting in clinical milk fever. Without treatment, levels may continue to decline to values as low as or even below 0.5 mmol/L (2 mg/dL), which is usually incompatible with life. Hypocalcemia is the cause of the signs of typical milk fever. Atony of skeletal muscle and plain muscle are well-known physiologic effects of hypocalcemia in ruminants. Experimental Hypocalcemia Hypocalcemia can be induced experimentally by administering Na2-EDTA intravenously, which results in the complex binding or chelation of ionized Ca. The IV infusion of Na2-EDTA into cows over a period of 4 to 8 hours results in severe hypocalcemia and paresis and has been used extensively as a model for the reproduction of the disease. A standardized flow rate of 1.2 mL/kg per hour of a 5% solution of Na2-EDTA until

recumbency results in changes in plasma ionized Ca, total Ca, inorganic phosphate (Pi), and magnesium comparable to what is observed in spontaneous cases of milk fever. Induced hypocalcemia results in depression of the frequency and amplitude of rumen contractions as early as 1.0 mmol/L of ionized serum Ca, well before any clinical signs of hypocalcemia are detectable and while feeding behavior and rumination are still normal. The induction of subclinical hypocalcemia in cows results in a linear decrease in feed intake and chewing activity as the plasma ionized calcium decreases. Feed intake depression was observed with ionized Ca concentrations below 0.9 mmol/L and before other signs commonly associated with hypocalcemia were recorded. Feed intake approached zero when ionized Ca concentrations declined to 0.6 mmol/L. This suggests that hypocalcemia may contribute to the reduction in feed intake prepartum and depresses the rumination process, ultimately leading to anorexia. Experimental hypocalcemia in cattle furthermore resulted in a marked reduction in cardiac stroke volume, a 50% reduction in arterial blood pressure, and a significant reduction in ruminal and abomasal tone and motility. In experimental hypocalcemia in sheep, blood flow is reduced by about 60% to all tissues except the kidney, heart, lung, and bladder, in which the reduction is not as high. During periods of prolonged hypocalcemia in cows and ewes, blood flow to skeletal muscles and the alimentary tract may be reduced to 60% to 70% of normal for a long period, which may present a predisposing factor for the downer-cow syndrome. Serum Ca and Pi levels are significantly lower in clinical cases than in comparable normal cows, and there is some relationship between the severity of the signs and the degree of biochemical change. Signs of hypocalcemic tetany, presumably attributable the increased membrane instability, commonly recognized in nonruminant species are observed in the initial stages of milk fever in cattle: • Nervousness and early excitement • Muscle twitching • Tetany, particularly of the hindlimbs • Hypersensitivity and convulsive movements of the head and limbs There are additional signs in the experimental disease, such as excessive salivation, excessive lip and tongue actions, and tail lifting. The serum muscle enzyme levels of creatine phosphokinase (CPK) and aminotransferase (AST) increase as a result of muscle injury associated with prolonged recumbency. Blood glucose levels increase, and serum Pi and potassium levels decrease. The prolonged infusion of Na2-EDTA in sheep over 18 hours at a rate to induce hypocalcemia and maintain recumbency resulted in prolonged periods of recumbency ranging from 36 to 64 hours before the animals were

1679

able to stand. There are also decreases in plasma sodium, plasma potassium, and erythrocyte potassium and prolonged increases in PCVs, suggesting that fluid replacement therapy may be indicated in cattle with prolonged recumbency associated with hypocalcemia. The activity of AST and CPK, the PCVs, and white blood cell (WBC) counts were elevated 24 hours later. Hypomagnesemia Hypomagnesemia is recognized as an importing contributing factor to periparturient hypocalcemia and has been proposed to be the major risk factor for milk fever occurring in cattle in mid- to late lactation.3 The two mechanism through which Mg deficiency may predispose to hypocalcemia are an impaired release of PTH in response to hypocalcemia and decreased tissue sensitivity to PTH in hypomagnesemic states.3 Hypomagnesemia can therefore predispose to clinical or subclinical milk fever by blunting the main counter-regulatory mechanism of hypocalcemia. Hypophosphatemia Low serum Pi concentrations are commonly observed in milk-fever cows, but also in healthy dairy cows, around parturition.7 Although the clinical relevance of hypo­ phosphatemia in recumbent cattle remains uncertain, empirical associations between hypophosphatemia and recumbency have been established.9 Anecdotal reports from field veterinarians suggest that the numbers of recumbent periparturient dairy cows not responding to standard therapy with intravenous Ca salts and showing pronounced hypophosphatemia have increased in recent years. To date, however, there is no unequivocal evidence corroborating the hypothesis that hypophosphatemia plays a role in periparturient recumbency or confirming the treatment efficacy of oral or parenteral Pi supplementation to recumbent cattle.7

CLINICAL FINDINGS Cattle Three stages of milk fever in cattle are commonly recognized and described. Stage 1 In the first stage, the cow is still standing. This is also the brief stage of nervousness, excitement, and tetany with hypersensitivity and muscle tremor of the head and limbs. The animal is disinclined to move and often has a decreased or no feed intake. There may be a slight shaking of the head, protrusion of the tongue, and grinding of the teeth. The rectal temperature is usually normal to slightly above normal; the skin may feel cool to the touch. The animal appears ataxic, with a stiff and insecure gait, and falls easily. Close examination reveals agalactia, rumen stasis, and scant feces. Heart rate and respirations may be within normal limits or slightly elevated.

1680

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Stage 2 The second stage is characterized by sternal recumbency with depressed consciousness; the cow has a drowsy appearance in sternal recumbency, usually with a lateral kink in the neck or the head turned into the flank (Fig. 17-4). When approached, some of these cows will open their mouths, extend the head and neck, and protrude their tongues, which may be an expression of apprehension and fear in an animal unable to stand. The tetany of the limbs present in the first stage is not present, and the cow is unable to stand. The muzzle is dry, the skin and extremities cool, and the rectal temperature subnormal (36 to 38° C, 97 to 101° F). There is a marked decrease in the absolute intensity of the heart sounds, whereas the heart rate is increased (about 80 bpm). The arterial pulse is weak and the venous pressure is also low, making it difficult to raise the jugular veins. The respirations are not markedly affected, although a mild forced expiratory grunt or groan is sometimes audible. Ruminal stasis and secondary bloat are common, and constipation is characteristic. There is also relaxation of the anus and loss of the anal reflex. The eyes are usually dry and staring. The pupillary light reflex is incomplete or absent, and the diameter of the pupil varies from normal to maximum dilatation. A detailed examination of the pupils of cows with parturient paresis, nonparetic disorders, and nonparturient paresis found that the mean sizes of the pupils were not significantly different from one another. Rather, disparity of the size of the pupils was common. In cows that develop hypocalcemia a few hours before or at the time of parturition, the second stage of parturition may be delayed. Vaginal examination usually reveals a fully dilated cervix and normal presentation of the

fetus. The cow may be in any stage of milk fever, and administration of Ca-salts IV will usually result in a rapid beneficial response and normal parturition. Prolapse of the uterus is a common complication of milk fever, and often the Ca levels are lower than in parturient cows without uterine prolapse. Thus it is standard practice to treat cases of uterine prolapse with IV calcium salts. Stage 3 The third stage is characterized by a severely obtunded or even comatose cow in lateral recumbency. There is complete flaccidity on passive movement, and the cow cannot assume sternal recumbency on its own. In general, the depression of temperature and the cardiovascular system are more marked. The heart sounds are almost inaudible, and the rate is increased up to 120 bpm; the pulse is almost impalpable, and it may be impossible to raise the jugular veins. Bloat is common because of prolonged rumen stasis and lateral recumbency. Without treatment, a few animals remain unchanged for several hours, but most become progressively worse during a period of several hours and die quietly from shock in a state of complete collapse. Concurrent Hypomagnesemia.  Mild to moderate tetany and hyperesthesia persisting beyond the first stage suggests a concurrent hypomagnesemia. There is excitement and fibrillary twitching of the eyelids, and tetanic convulsions are readily precipitated by sound or touch. Trismus may be present. The heart and respiratory rates are increased, and the heart sounds are much louder than normal. Without treatment, death occurs during a convulsion.

Fig. 17-4  Friesian cow with stage 2 periparturient hypocalcemia. The cow is unable to stand without assistance.

Sheep and Goats The disease in pastured ewes is similar to that in cattle. The early signs include a stilty, proppy gait and tremor of the shoulder muscles. Recumbency follows, sometimes with tetany of the limbs, but the proportion of ewes with hypocalcemia that are recumbent in the early stages is much less than in cattle. A similar generalization applies to female goats. The characteristic posture is sternal recumbency, with the legs under the body or stretched out behind. The head is rested on the ground, and there may be an accumulation of mucus exudate in the nostrils. The venous blood pressure is low and the pulse impalpable. Mental depression is evidenced by a drowsy appearance and depression of the corneal reflex. There is loss of the anal reflex, constipation, tachycardia, hyposensitivity, ruminal stasis and tympany, salivation, and tachypnea. Response to parenteral treatment with Ca salts is rapid; the ewe is normal 30 minutes after an SC injection. Death often occurs within 6 to 12 hours if treatment is not administered. The syndrome is usually more severe in pregnant than in lactating ewes, possibly because of the simultaneous occurrence of pregnancy toxemia or hypomagnesemia. Fat late-pregnant ewes on high-grain diets indoors or in feedlots show a similar syndrome accompanied by prolapses of the vagina and intestine.

CLINICAL PATHOLOGY

Total serum Ca levels are reduced to below 2.0 mmol/L (8 mg/dL), usually to below 1.2 mmol/L (5 mg/dL), and sometimes to as low as 0.5 mmol/L (2 mg/dL). The reduction is usually, but not always, proportional to the severity of the clinical syndrome. Average figures for total serum Ca levels in the three species are as follows: cows, 1.30 ± 0.30 mmol/L (5.2 ± 1.2 mg/dL); ewes, 1.15 ± 0.37 mmol/L (4.6 ± 1. 5 mg/dL); goat does, 0.94 ± 0.15 mmol/L (3.8 ± 0.6 mg/dL). Although the concentration of ionized Ca, which is the biologically active fraction of the total Ca pool, is the factor determining the presence and severity of clinical signs in hypocalcemic animals, the total serum Ca concentration is commonly used for convenience. Measurement of ionized Ca concentration requires the use of ion-selective electrodes, which have become much more accessible in recent decades. Nonetheless, the association between ionized and total Ca in serum is tight, with excellent correlation between the two, which is why total Ca concentration in serum is considered clinically useful and sufficiently accurate in practice.3 Between 42% and 48% of the total Ca content in the extracellular space is available as biologically active ionized Ca. A decrease in serum albumin or acidemia tends to increase the ionized Ca fraction, whereas alkalemia or an increase in serum albumin tends to decrease the proportion of ionized Ca.3 Equine, bovine, and ovine blood may be

Metabolic Diseases of Ruminants

VetBooks.ir

stored for up to 48 hours without any clinically relevant alteration of blood Ca ion concentration. Levels of ionized Ca in the venous whole blood of cows are as follows: normal, 1.06 to 1.26 mmol/L (4.3 to 5.1 mg/dL); slight hypocalcemia, 1.05 to 0.80 mmol/L (4.2 to 3.2 mg/ dL); moderate, 0.79 to 0.50 mmol/L (3.2 to 2.0 mg/dL); severe hypocalcemia, less than 0.50 mmol/L ( urine > saliva. Skeletal Muscle Potassium Content Skeletal muscle potassium concentration is considered the most sensitive and specific method for assessing whole-body potassium status and therefore provides the goldstandard test. Skeletal muscle is considered the best tissue to sample because it contains approximately 75% of the whole-body stores of potassium. A standardized muscle should be evaluated in cattle because differences in potassium content of greater than 15% are present in individual animals, and this muscle-to-muscle variation is greater than that produced by breed. Plasma Potassium Concentration Determination of serum/plasma potassium concentration is required to confirm a suspected diagnosis of hypokalemia. A serum potassium concentration less than 2.5 mEq/L reflects severe hypokalemia, and most animals will be weak or recumbent. A serum potassium concentration of 2.5 to 3.5 mEq/L reflects moderate hypokalemia, and some cattle will be recumbent or appear weak, with depressed gastrointestinal motility. In addition to measurement of serum potassium concentration, determination of the serum concentrations of sodium, chloride,

1692

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

calcium, and phosphorus and the serum activities of creatine kinase and aspartate aminotransferase can be very helpful in guiding treatment of cattle with hypokalemia. Serum potassium concentration is usually a little higher than plasma potassium concentration because platelet activation releases potassium. In summary, a serum/ plasma potassium concentration below the normal range provides unequivocal evidence of hypokalemia unless there is concurrent hyperinsulinemia or alkalemia.4,5 However, because more than 95% to 98% of wholebody potassium stores are intracellular, it is likely that serum/plasma potassium concentration is not as sensitive as skeletal muscle potassium content in indicating whole-body potassium depletion. Milk Potassium Concentration Milk potassium concentration is theoretically more sensitive than serum/plasma potassium concentration in detecting wholebody potassium depletion in individual cows because the milk concentration of potassium is constant for an individual cow. Potassium depletion in lactating dairy cows caused milk potassium concentration to decrease from 1.45 g/L to 1.28 g/L; this was a greater percentage decrease than that in the plasma or whole blood of cattle with whole-body potassium depletion. However, there is marked individual variation in the milk concentration of potassium in healthy cattle, with variations of up to 50% occurring between cows. This variability appears to be a result of changes in milk fat, protein, and lactose percentage, with the highest correlation of milk potassium concentration being with milk lactose concentration (R = −0.53 or −0.74). The relationship between potassium and lactose is attributable to the fact that these are important contributors to milk osmolality, which is constant and isotonic. Milk potassium concentration also changes during lactation, being 42 mmol/L in early lactation, 40 mmol/L in midlactation, and 27 mmol/L in late lactation, with a mean bulk milk potassium concentration of 37 mmol/L. The large cow-to-cow variability in milk potassium concentration and dependence on milk lactose concentration make it difficult to produce a suitable cut-point for identifying whole-body potassium depletion in sick lactating dairy cows. However, milk potassium concentration has clinical utility in monitoring potassium homeostasis over time in an individual cow. Erythrocyte Potassium Concentration Erythrocyte potassium concentration is determined by measuring plasma potassium concentration and hematocrit, and then adding sufficient distilled water to hemolyze the erythrocytes followed by potassium measurement of the hemolyzed fluid and mathematical calculation. There is marked cow-to-cow variability in the erythrocyte

potassium concentration (7 to 70 mmol/L) and sodium concentration (15 to 87 mmol/L) of healthy cattle that has a genetic basis with no breed influence. There are two main peaks of cellular potassium concentration, one at 20 mmol/L and a second at 50 mmol/L. In lactating dairy cattle with induced wholebody potassium deficiency, whole-blood potassium concentration changed similarly to plasma potassium concentration. However, in 180 cows, no relationship between plasma potassium concentration and erythrocyte potassium concentration was found. Measurement of erythrocyte or whole-blood potassium concentration is not currently recommended in evaluating whole-body potassium status. Urine Potassium Concentration Urine potassium concentrations are normally high (454 ± 112  mEq/L) but variable, with a mean fractional clearance of 82% and a coefficient of variation of 61%. The large variability in urine potassium concentration makes it difficult to produce a suitable cut-point for identifying whole-body potassium depletion. However, determination of urine potassium concentration has clinical utility in an individual cow ingesting a constant diet over time because it reflects potassium homeostasis. Urine pH may provide some value as a better screening test because aciduria may be present in response to a marked decrease in urine potassium concentration.10 Salivary Potassium Concentration Salivary potassium concentrations are more influenced by aldosterone in the response to changes in serum sodium concentration, and salivary potassium concentration must therefore be compared with the salivary sodium concentration (one-for-one exchange), sodium homeostasis, and the ratio of serum sodium to potassium to have clinical utility. The normal saliva potassium concentration shows a large range of 4 to 70 mEq/L, with sodium homeostasis having the greatest effect. A study in cattle with left-displaced abomasum, right-displaced abomasum, or abomasal volvulus indicated no association between salivary potassium concentration and serum potassium concentration. Taken together, it appears that measurement of salivary potassium concentration provides minimal insight into whole-body potassium status.

NECROPSY FINDINGS

Necropsy of cattle with hypokalemiainduced recumbency reveals the presence of muscle necrosis in the pelvic limbs. Histologic examination of non-weight-bearing muscles reveals multifocal myonecrosis with microphage infiltration and myofiber vacuolation, which is characteristic of hypokalemic myopathy in humans and dogs. It is important to note that hypokalemic

myopathy is also present in muscles not subject to ischemia of recumbency.

TREATMENT

Treatment of hypokalemia in lactating dairy cows should focus on surgical correction of abomasal displacement, increasing the potassium intake by increasing dry matter intake or the oral administration of KCl, and correction of hypochloremia, alkalemia, metabolic alkalosis, and dehydration.2 Oral potassium administration is the method of choice for treating hypokalemia. Inappetent adult cattle should initially be treated with 120 g of KCl PO, followed by an additional oral treatment of 120 g KCl 12 hours later, for a total 24-hour treatment of 240 g KCl (0.4 g/kg BW).6 Higher oral doses of KCl are not recommended because they can lead to diarrhea, excessive salivation, muscular tremors of the legs, and excitability. Potassium is rarely administered intravenously; the IV route is used only for the initial treatment of recumbent ruminants with severe hypokalemia and rumen atony because it is much more dangerous and expensive than oral treatment. The most aggressive IV treatment protocol is an isotonic solution of KCl (1.15% KCl), which should be administered at less than 3.2 mL/ kg/hr, equivalent to a maximal delivery rate of 0.5 mEq of potassium/kg BW per hour. Higher rates of potassium administration run the risk of inducing hemodynamically important arrhythmias, including ventricular premature complexes that can lead to ventricular fibrillation and death. Palatable hay and propylene glycol orally are recommended. In a series of 14 cases, treatment consisted of potassium chloride given intravenously and orally at an average total daily dose of 0.42 g/kg BW (26 g orally and 16 g IV) for an average of 5 days, resulting in recovery in 11 cases after an average of 3 days. During recumbency, affected cattle require special attention to minimize ischemic necrosis of muscles of the pelvic limbs. Glucocorticoids are often used to treat ketosis, and the most commonly used glucocorticoids are dexamethasone and isoflupredone acetate. Dexamethasone has little mineralocorticoid activity compared with prednisone and prednisolone, which are related chemically to isoflupredone. Dexamethasone is recommended for the treatment of ketosis in dairy cattle at a single dose of 10 to 20 mg IM, and repeated, if necessary, 12 to 24 hours later. Field observations indicate that repeated doses of isoflupredone acetate decrease plasma concentrations of potassium by 70% to 80%, which suggests a strong mineralocorticoid activity. It is recommended that isoflupredone acetate be used judiciously and animals be monitored for plasma potassium and any evidence of weakness and recumbency. Treatment with oral potassium chloride may be required, but treatment may be ineffective.

Metabolic Diseases of Ruminants

CONTROL

VetBooks.ir

Oral administration of potassium is a mandatory component of fluid and electrolyte administration to inappetent lactating dairy cattle. Ensuring an adequate dry matter intake is the best method for preventing hypokalemia in lactating dairy cattle. TREATMENT AND CONTROL Treatment KCl 120 g PO, followed by an additional oral treatment of 120 g KCl 12 hours later, for a total 24-hour treatment of 240 g KCl (0.4 g/kg BW) (R-1) KCl 1.15% IV, less than 3.2 (mL/kg BW)/hour, equivalent to a maximal delivery rate of 0.5 mEq of potassium/kg BW per hour (R-2) Isoflupredone acetate (R-4) Control Maintain adequate dry matter intake (R-1)

FURTHER READING

Constable PD. Fluids and electrolytes. Vet Clin North Am Food Anim Pract. 2003;19(3):1-40. Sattler N, Fecteau G. Hypokalemia syndrome in cattle. Vet Clin North Am Food Anim Pract. 2014;30:351-357.

REFERENCES

1. Constable PD, et al. J Dairy Sci. 2009;92(1):296. 2. Constable PD, et al. J Am Vet Med Assoc. 2013;242:826. 3. Coffer NJ, et al. Am J Vet Res. 2006;67:1244. 4. Grünberg W, et al. J Am Vet Med Assoc. 2006;229:413. 5. Grünberg W, et al. J Vet Intern Med. 2006;20:1471. 6. Constable PD, et al. J Dairy Sci. 2014;97(3):1413. 7. Greenlee M, et al. Ann Intern Med. 2009;150:619. 8. Türck G, Leonhard-Marek S. J Dairy Sci. 2010;93(8):3561. 9. Zurr L, Leonhard-Marek S. J Dairy Sci. 2012;95:5750. 10. Constable PD, et al. Am J Vet Res. 2009;70(7):915.

DOWNER-COW SYNDROME SYNOPSIS Etiology Ischemic myopathy of large muscles of pelvic limbs and ischemic neuropathies of obturator or sciatic nerve or its branches secondary to prolonged recumbency associated with milk fever or dystocia; injury of bones, joints, and muscles; undetermined etiologies. Epidemiology Most common in dairy cows with previous episodes of milk fever; in beef cows after prolonged or difficult calving. Delay of more than 4 hours in treatment for recumbent milk-fever cows. Hypophosphatemia and/or hypokalemia have been discussed as potential risk factors. Signs Alert downer cows: Unable to stand following treatment for milk fever. Sternal recumbency; normal mental status, vital signs, and alimentary tract. Appetite and thirst normal or mildly decreased. Most will

stand in few days if provided good clinical care and secondary muscle necrosis is minimized. Nonalert downer cows: Persistent recumbency with altered mentation and vital signs; frequently unable to maintain sternal recumbency; abnormal position of legs; groaning; anorexia; die in several days. Clinical pathology Increased serum activity of creatine kinase (CK) and aspartate aminotransferase (AST); serum phosphorus and potassium concentrations may be subnormal or elevated; proteinuria, myoglobinuria. Necropsy findings Ischemic necrosis, edema and hemorrhage of large medial thigh muscles. Diagnostic confirmation Increased serum activities of CK, AST, proteinuria; myoglobinuria necropsy lesions in cow unable to rise with no other lesions. Treatment Provide excellent bedding or ground surface such as sand or dirt pack. Roll animal from side to side every few hours. Antiinflammatory therapy/pain management. Fluid and electrolyte therapy as necessary. Hoist cows making attempts to stand. Control All recently calved dairy cows that are at high risk for milk fever must be observed closely 12 to 24 hours before and after calving for evidence of milk fever and while still standing; if recumbent, do not delay treatment for more than 1 hour. Can treat all high-risk cows with calcium salts orally to prevent clinical milk fever.

The term downer cow first appeared in the veterinary literature in the 1950s and referred to cattle that were too injured, weak, or sick to stand or walk without assistance.1 In most of the early publications using this terminology a case definition was not provided or was imprecise, such as “cattle unable to rise” or “unable to stand without assistance,” and did not make reference to possible etiologies, duration of recumbency, or outcome.2 More recently the term downer cow was used to denote nonambulatory cattle recumbent for at least 24 hours without obvious reason.1 A further classification of downer cows into mentally alert, nonambulatory cattle that are able to maintain themselves in sternal recumbency, so-called alert downer cows, and cows with moderate to severe mental obtundation and abnormal vital signs that frequently are unable to maintain sternal recumbency, the so-called nonalert downer cows, was proposed.3 The term creeper cows is sometimes used to denote alert recumbent cows that are unable to bear weight on their hindlimbs but that use the forelegs to propel themselves over short distances.

ETIOLOGY

Alert downer cows are in most cases recumbent because of musculoskeletal or neuro-

1693

logic injuries such as lesions of the sciatic or obturator nerve secondary to dystocia (calving paralysis), fractures of long bones or the pelvis, hip luxation, or muscle injury as a result of primary trauma or secondary to prolonged recumbency. Nonalert downer cows comprise animals with systemic disease affecting mental status and general attitude, such as periparturient hypocalcemia, septicemia, hypovolemia, diffuse peritonitis, and severe hepatic lipidosis, or neurologic diseases affecting the brainstem or cortex.3 In most cases, the downer-cow syndrome is a complication of milk fever. Myopathies and neuropathies develop in nonambulatory cows secondary to prolonged periods of recumbency. Ischemic myopathy affecting the large muscles of the pelvic limbs and injuries to the tissues around the hip joint and of the obturator muscles are common in cows that do not fully recover and stand. Injuries to the musculoskeletal system are also common as a result of cows “spreadeagling” their hindlimbs if they are unsteady during parturition or forced to stand or walk on a slippery floor immediately before or following parturition. A survey conducted among dairy operations in 21 U.S. states determined that the three most common causes for persistent recumbency were periparturient hypocalcemia (19%), calving-related injuries (22%), and injuries from slipping or falling (15%). Beef cattle operations reported calving paralysis as the single most common cause for downer-cow syndrome.1

EPIDEMIOLOGY Occurrence The disease is most common in dairy cows and typically occurs within the first 2 or 3 days after calving, often immediately following an episode of milk fever. Other debilitating conditions of periparturient cows that can be associated with persistent recumbency include acute coliform mastitis, septic metritis, and acute rumen acidosis (grain overload). In the United States an estimated 270,000 cattle became nonambulatory on-farm in 2004, of which 57.4% were dairy and 31.5% beef cattle, corresponding to 1.2% of dairy cattle and 0.2% of beef cattle becoming persistently recumbent in 1 year.4 An older survey conducted in 1986 in Minnesota that included data from 738 dairy operations and 34,656 cow years at risk reported incidences per herd and year between 0.4% and 2.1% (case definition in this study: “sternal recumbency for at least 24 h for no obvious reason”). The overall outcome was that 33% of downer cows recovered, 23% were slaughtered, and 44% died. The owners perceived that downer cows were high producers (48%) or average producers (46%), with only 6% being low producers. Approximately 58% of cases occurred within 1 day of parturition, and 37% occurred during the first 100 days

1694

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

of lactation. The incidence was highest (39%) during the three coldest months: December to February. A clinical survey conducted in New Zealand and including 433 periparturient recumbent cows reported a recovery rate of 39%, whereas 30% died, and 32% had to be destroyed. The case-fatality rate in this study was 11% higher in precalving recumbent cows than postcalving cows. A 2006 survey including dairy operations from 21 U.S. states reported that 78.6% of participating operations had at least one downer cow in 2006.2 The case definition of a downer cow in this study was “nonambulatory cattle that were unable to stand for any length of time, including those that recovered.”2 Because it is a syndrome lacking in clinical definition and includes all those “other cases” that cannot be otherwise classified, downer-cow incidence varies depending on the clinical acuity of the individual veterinarian and various environmental factors in different areas. In any case, the incidence seems to be increasing, particularly in intensive dairy farming areas, although this impression could arise from the increased necessity to effect a cure in valuable animals. Risk Factors Animal Risk Factors Complication of Milk Fever.  Prolonged recumbency after an episode of clinical milk fever either because of a delay in administration of proper treatment or delayed response to treatment is considered the most common primary cause of downer-cow syndrome. The incidences for downer-cow syndrome that is associated with milk fever reported in the literature range from 3.8% to 28.2% of all milk-fever cases.5 Prolonged recumbency, regardless of the primary cause, results in increased tissue pressure over a confined anatomic area, causing local ischemia and neuromuscular dysfunction. An insecure gait of a hypocalcemic periparturient cow presents an increased risk of injury from slipping or falling, such as muscle rupture, bone fractures, or hip luxation, that can result in downer-cow syndrome. Traumatic Injuries to Pelvis and Pelvic Limbs.  Traumatic injuries to bones, muscles, and nerves can be directly related to parturition (e.g. calving paralysis), be associated with muscle weakness and an insecure gait (e.g. in hypocalcemic cattle), or be the result of an inadvertent accident. Calving paralysis refers to a paresis or paralysis of one or both hindlimbs caused by a lesion of the obturator nerve and/or the lumbar root of the sciatic nerve inflicted during the calving process. Both nerves are vulnerable to compression between the bony birth canal and the calf at parturition; accordingly, nerve damage is most commonly diagnosed after dystocias, deliveries of large calves, or prolonged calvings. Calving paralysis is con-

sidered the most common cause for persistent recumbency in beef cattle. Pressure injuries of the superficial nerves of the extremities may occur as secondary lesions in cows that are recumbent for an unrelated reason. Serum Electrolyte Imbalances.  Apart from hypocalcemia, hypophosphatemia, hypokalemia, and hypomagnesemia have been incriminated as potential factors contributing to downer-cow syndrome. Hypophosphatemia is a common finding in recumbent but also in healthy periparturient cows;6 it is the mineral imbalance most commonly quoted as risk factor, especially in the socalled creeper cows, which are bright and alert and crawl about, but are unable to rise. The clinical relevance of hypophosphatemia in persistently recumbent animals has been debated contentiously, but an undisputed empirical observation is that hypophosphatemia is more common or more pronounced in recumbent periparturient cows that are unresponsive to intravenous calcium administration at least in very early stages of recumbency.5,6 However, the mechanisms through which phosphate depletion may cause persistent recumbency are not well understood, and treatment response to oral or parenteral administration of phosphate salts is inconsistent.6 Studies including cattle that were nonambulatory for longer than just a few hours, in contrast, found that low serum phosphorus levels are suggestive of a good prognosis, whereas nonsurvivors tend to have higher serum phosphorus concentrations.3 A likely explanation for this finding is that cows recumbent for longer time periods may have developed more severe muscle damage that is associated with release of intracellular phosphorus into the circulation and thus an increase of the serum phosphorus concentration. A long-term low-level hypomagnesemia has been associated with downer-cow syndrome, especially when it accompanies hypocalcemia. But it is usually manifested by a tetanic hyperesthetic state, which is not part of downer-cow syndrome. Severe hypokalemia in cattle is associated with signs of depression and profound skeletal muscle weakness leading to recumbency.7 Reports of pronounced hypokalemia in individual animals that were associated with persistent recumbency, with serum potassium concentrations below 2.0 mmol/L, have accumulated over the past decades. These cases have in most instances been traced back to the repeated use of isoflupredone, a mineral corticoid with strong kaliuretic effect that was commonly used for the treatment of ketosis in the United States.7 Mild to moderate hypokalemia is known to occur in early lactating and anorectic cows, but the role of this mild form in the pathogenesis of downer-cow syndrome needs to be determined.8

The age and stage of lactation of a recumbent cow were found to be risk factors for nonrecovery. Recovery rates of nonambulatory cows were 10.1% for first-lactation cows, 17.7% for cows in their second to fourth lactation, and 22.2% for fifth-lactation cows.2 Cows less than 15 days in milk had a recovery rate of 28.4%, whereas cows in later lactation had a 6.2% chance of making a full recovery.2 Higher recovery rates in older cows and cows that were earlier in lactation have been attributed to an association between persistent recumbency and hypocalcemia. Cows that are older and earlier in lactation are more likely to be recumbent because of hypocalcemia as primary or contributing cause, which has a better prognosis than persistent recumbency for other reasons. Duration of recumbency was also found to be associated with the likelihood of making a full recovery. Cows that were down for less than 24 hours recovered in 32% of the cases, whereas cows recumbent for longer periods had an 8.2% chance of recovery.2 A high body-condition score is a recognized risk factor for milk fever and therefore must also be considered as predisposing for downer-cow syndrome. Cows with a BCS above 4.0/5 around calving were found to be at 4.3 times higher risk to become nonambulatory than thinner cows. In contrast, cows in poor body condition, with a BCS below 2.5/5, recovered in 8.1% of all cases, whereas 16.6% of cows with a BCS of 2.75 or higher made a full recovery.2 Environmental and Management Risk Factors A slippery ground surface is a major risk factor. Cattle that must walk across slippery floors, especially at the time of calving, may slip and fall and injure the large muscles of the pelvic limbs, resulting in an inability to stand.

PATHOGENESIS

In most cases downer-cow syndrome is a complication of an unrelated primary problem causing muscle weakness or persistent recumbency. Primary conditions that can lead to downer-cow syndrome have been grouped into four major categories: metabolic disorders (e.g., hypocalcemia, hypokalemia), acute systemic illness (e.g., coliform mastitis, toxic metritis), musculoskeletal disorders (e.g., fractures, joint luxation), and undetermined causes.9 Prolonged recumbency will result in secondary damage from excessive pressure on limbs squeezed between the body and the ground or from struggling to get up. If severe enough, these secondary lesions may prevent the affected cow from getting up, even though the primary cause of recumbency may in the meantime be resolved. Secondary damage can affect muscles, nerves, or other structural components such as bones or joints. Regardless of the initial cause,

Metabolic Diseases of Ruminants

VetBooks.ir

prolonged recumbency results in varying degrees of pressure damage predominantly affecting the hindlimbs. Based on the results of experimental studies, it has been suggested that 6 hours of recumbency is the time threshold, beyond which tissue damage as a result of excessive weight bearing must be expected. This underscores the importance of handling any persistently recumbent cow as a medical emergency. Pressure damage in recumbent cattle primarily occurs in the major muscles of the hindlimbs, particularly the semitendinous muscle, muscles caudal to the stifle, and the peripheral sciatic nerve and its branches. The local tissue damage is referred to as compartment syndrome; systemic effects resulting from local tissue damage are summarized in the so-called crush syndrome. Compartment Syndrome A compartment of the body is composed of muscle and nerves within an anatomically defined area that is surrounded by a rigid muscle fascia layer. In a recumbent cow, the compartments of interest are the ones of the upper part and to a lesser extent the lower part of each hindlimb. The initial pressure acting on the hindlimb located underneath the body of a recumbent cow depends on the body weight resting on this limb and the rigidity of the ground on which the cow is lying. This pressure on the limb directly translates into increased pressure within the affected compartment and will result in partial or complete occlusion of venous blood flow before the arterial blood flow of the affected region is decreased. The mismatch between blood flow into and out of the compartment leads to a further pressure increase within the compartment. Impaired blood supply to muscles and nerves and ensuing tissue hypoxia will add to the direct damage from mechanical compression. The thick muscle-fascial boundaries surrounding the compartment prevent tissue expansion that would relieve the structures within the compartment from the excessive pressure. Cell damage and inflammation are associated with swelling, causing a further increase in pressure and contributing to a detrimental cascade of events. Experimental external compression of the pelvic limb in goats, to simulate limb compression in recumbent cows, resulted in a marked reduction in the nerve conduction velocity of the peroneal nerve, which was associated with clinically evident limb dysfunction. Crush Syndrome Crush syndrome refers to the sum of the systemic effects of extensive muscle tissue injury and is attributed to the massive release of muscle-tissue breakdown products into the blood circulation. Notably, a large increase in the serum activity of muscle enzymes, such as aspartate aminotransferase (AST) or

creatine kinase (CK); increases in serum concentration of predominantly intracellular electrolytes, such as potassium and phosphorus; and ultimately the appearance of myoglobin in urine are indicative of crush syndrome. Myoglobinuria is a potentially life-threatening complication of downer-cow syndrome that can lead to acute renal failure. Experimental Sternal Recumbency Experimentally induced sternal recumbency with one hindlimb positioned under the body to simulate prolonged recumbency will result in a swollen rigid limb within 6 to 9 hours. Following injury to the muscle cells, the serum activity of CK is markedly elevated at about 12 hours after the onset of recumbency. Proteinuria and in some severe cases myoglobinuria occur between 12 and 36 hours after the onset of prolonged recumbency, as a result the release of myoglobin from damaged muscles. In cows that make efforts to stand but cannot do so, continued struggling may result in rupture of muscle fibers and hemorrhage. Acute focal myocarditis occurs in about 10% of cases, resulting in tachycardia, arrhythmia, and the unfavorable response to IV calcium salts observed in some cases. The cause of the myocardial lesion is unknown, but repeated administration of calcium salts has been suggested. The prolonged recumbency can result in additional complications, such as acute mastitis, decubitus ulcers, and traumatic injuries of the limbs. The pathogenesis of the nonalert downer cow is not understood. Most such cows have had an initial episode of milk fever and did not respond satisfactorily. Within 1 or 2 days, affected cows have a preference for lateral recumbency and exhibit expiratory moaning and groaning. They represent about 2% of all cases of milk fever.

CLINICAL FINDINGS

Downer-cow syndrome may occur independently or follow apparent recovery after treatment for milk fever, except for the prolonged recumbency. In the typical case, affected cows either make no effort or are unable to stand following treatment for parturient paresis. About 30% of cows treated for milk fever will not stand for up to 24 hours following treatment. Affected cows are usually bright and alert with good or only mildly depressed feed intake and are thus classified as alert downer cows. The temperature is normal and the heart rate may be normal or elevated to 80 to 100 bpm. Tachycardia and arrhythmia occur in some cows, especially immediately following the administration of IV calcium, and sudden death has occurred. Respirations are usually unaffected. Defecation and urination are normal, but proteinuria is common and may indicate extensive muscle damage if marked. Some affected cows may make no effort to stand. Others will make frequent attempts

1695

to stand but are unable to fully extend their pelvic limbs and lift their hindquarters more than 20 to 30 cm from the ground. On a nonslippery surface (bare ground, sand pack, or deep bedding) some cows are able to stand with some assistance by lifting on the tailhead or with the use of hip slings. Those cows that do not make an effort to stand usually cannot stand even with assistance, and if supported with cow slings, they will usually make no effort to bear weight with either the hindlimbs or the forelimbs. Their limbs appear stiff, painful, or numb, and they are unable or reluctant to bear weight. Damage to the peroneal nerve is usually present when there is hyperflexion of the fetlock joints, which is evident if and when the cow is able to stand and bear weight on the hindlimbs. In some cases, the hindlimbs are extended on each side of the cow and reach up to the elbows on each side. In this position, the cow is bearing considerable weight on the medial thigh musculature and causing ischemic myopathy. This abnormal position of the legs may also result from dislocation of one or both hip joints or be associated with traumatic injuries surrounding the hip joints with or without rupture of the ligamentum teres. Regardless of the cause, the cow prefers this leg position and invariably will shift the legs back to the abnormal position if they are placed in their normal position. In some cows, the signs may be more marked and bizarre, including a tendency to lie in lateral recumbency with the head drawn back. When placed and propped up in sternal recumbency, these cows appear almost normal, but when they are left alone, they revert to the position of lateral recumbency within a short period of time. Still more severe cases are hyperesthetic, and the limbs may be slightly stiff, but only when the cow is lying in lateral recumbency. These severe cases do not usually eat or drink and have been described as nonalert downers. Complications in downer-cow syndrome are common and often result in death or the need for euthanasia. Coliform mastitis, decubitus ulceration, especially over the prominences of the hock and elbow joint, and traumatic injuries around the tuber coxae caused by the hip slings are common. When these complications occur in the early stages of the disease, they commonly interfere with any progress being made and become the focus of clinical attention. The course of the disease is variable and dependent on the nature and extent of the lesions and the quality of the care and comfort that is provided for the cow during the first few days. About 50% of downer cows will stand within 4 days or less if cared for properly. The prognosis is poor for those that are still recumbent after 7 days, although some affected cows have been down for 10 to 14 days and subsequently stood up and recovered. Death may occur in 48 to 72

1696

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

hours following the onset and is usually associated with myocarditis. Clinical Examination of the Downer Cow Clinical examination of the downer cow can be very difficult and challenging depending on the environmental circumstances and the physical size of the animal.9 Causes of persistent recumbency in cattle include metabolic, musculoskeletal, neurologic, neoplastic, and inflammatory disease; accordingly, obtaining an adequate history and conducting a thorough physical examination are indispensable.10 Key aspects of the history include age and stage of lactation of the animal, duration of recumbency, any previous clinical abnormalities before the recumbent stage, any previous treatments, diet and accidental access to new feeds, sudden unaccustomed exercise, and assessment of the management provided. The environment and the ground surface surrounding the recumbent animal may provide clues about the possibility that the animal slipped, fell, and was injured. A systematic physical examination of all accessible body systems is necessary. The animal should be examined visually from a distance for evidence of abnormalities of the carriage of the head and neck, to observe the position of the limbs, and to observe any attempts of the animal to stand or creep along the ground surface. The details of the clinical examination are presented elsewhere in this book. The standard close clinical examination is necessary to determine body temperature, heart rate and pulse, respiratory rate, and the state of the major body systems, such as the respiratory tract, cardiovascular system, central nervous system for mental state, gastrointestinal tract, mammary gland, and reproductive tract, any of which may indicate the presence of abnormalities associated with shock that results in recumbency. In the recently calved cow, particular emphasis must be given to adequate examination of the udder for mastitis, the uterus for metritis, and the gastrointestinal tract for diseases associated with toxemia, dehydration, and shock (abomasal volvulus, acute diffuse peritonitis, carbohydrate engorgement) that result in recumbency. A urine sample must always be obtained and tested for ketones and the presence of myoglobinuria. Careful systematic examination of the musculoskeletal system includes palpating the muscles, bones, joints, and feet of each limb, including passive flexion and extension of each limb. The coxofemoral joints are examined for evidence of dislocation. The vertebral column is examined for evidence of fracture or dislocation of vertebrae. It is important to examine both sides of the animal, which means rolling the cow over from side to side; often the animal may have to be rolled over more than once to repeat a particular examination.

A neurologic examination includes examination of the withdrawal reflexes, patellar reflexes, and sensation of all four limbs and the reflex arcs of the spinal cord; careful examination of lumbar and sacral areas, including sensation and tone in the tail; and examination of the cranial nerves. The examination can be extended by lifting the downer cow with appropriate lifters and observing if the animal extends its limbs and attempts to bear weight. While the animal is being assisted to stand, additional examinations of other parts of the body can be made.

CLINICAL PATHOLOGY

The serum calcium and glucose concentrations are frequently within the normal range, whereas phosphorus and potassium concentrations may be decreased in cows with depressed feed intake or increased in animals with more pronounced muscle damage and/ or dehydration. Results of hematologic examinations are usually unremarkable in early stages of the recumbency. The serum activity of CK and AST are usually markedly elevated by 18 to 24 hours after the onset of recumbency. Very high levels of serum CK activity shortly after the onset of recumbency that decline markedly within the following 24 to 48 hours are indicative of an acute muscle trauma (e.g., muscle rupture) that may be the cause of recumbency. More moderate elevation of the serum activity of CK with a tendency to slightly increase or remain constant over the following days is suggestive of continuous and ongoing muscle trauma resulting from prolonged muscle-tissue compression. Muscle tissue is rich in CK. and the plasma half-life of this enzyme in cattle is only about 8 to 9 hours; this parameter is therefore a sensitive but short-lived marker of muscle damage. When interpreting the serum CK activity of a recumbent cow, it is critical to consider the time of sample collection relative to onset of recumbency. AST, in contrast, has a considerably longer half-life and remains elevated for several days after initial trauma. In a series of 262 recumbent dairy cows, serum samples were analyzed for CK, lactate dehydrogenase (LDH), and AST to evaluate the value of serum enzyme activities for predicting a failure to recover. The optimal cutoff points maximizing the sensitivity and specificity of the tests were 2330, 2225, and 171 U/L for CK, LDH, and AST, respectively. The predictive value of AST was significantly better, with optimal cutoff points of 128 and 189 U/L, respectively. AST provided the best predictive indicator of whether a recumbent cow would not recover, the best results being obtained with serum samples taken on the first day of recumbency. In experimentally induced recumbency in cows, the CK activity remained within normal limits for the first 6 hours. However, by 12 hours there was a marked increase to mean

values of 12,000 U/L rising to 40,000 U/L by 24 hours. There may be moderate ketonuria. A marked proteinuria is usually evident by 18 to 24 hours after the onset of recumbency. The proteinuria may persist for several days or be absent within a few days. In severe cases, the urine may be brown and turbid because of severe myoglobinuria. Elevations of serum urea, muscle enzymes, and laboratory evidence of inflammation are considered the best prognostic indicators of an unfavorable recovery. The recovery rate was lower in cows with a total protein : fibrinogen ratio less than 10 : 1, and evidence of neutropenia and/or left shift. Cows with a serum urea level above 25 mmol/L and serum creatinine levels above 130 mmol/L had a poor prognosis.

NECROPSY FINDINGS

Hemorrhages and edema of the skin of traumatic origin are common. The major pathologic changes consist of hemorrhages and degeneration of the medial thigh muscles. Hemorrhages around the hip joint with or without rupture of the ligamentum teres are also common. Local areas of ischemic necrosis of the musculature (gracilis, pectineus, and adductor muscles) occur at the anterior edge of the pelvic symphysis. Eosinophilic infiltration of ruptured necrotic thigh muscles of downer cows has been described. Hemorrhages and edema of the nerves of the limbs (obturator, sciatic, peroneal, radial) are also common and usually associated with severe muscle damage. The heart is dilated and flabby; histologically, there is focal myocarditis. There is fatty degeneration of the liver, and the adrenal glands are enlarged. Histologically, there are also degenerative changes in the glomerular and tubular epithelium of the kidneys.

DIFFERENTIAL DIAGNOSIS The diagnosis of downer-cow syndrome is typically made by exclusion of all other known causes of recumbency in a cow persistently recumbent for at least 24 hours while having received two courses of parenteral calcium treatment. Differential diagnoses for alert downer cows: • Hypocalcemia • Calving paralysis • Fractures of bone or pelvis • Hip luxation • Hypokalemia • Botulism • Spinal lymphosarcoma (BLV) Differential diagnoses for nonalert downer cows: • Hypokalcemia • Hepatic lipidosis/puerperal liver coma • Coliform mastitis • Toxic metritis

Metabolic Diseases of Ruminants

VetBooks.ir

• • • • • • • • • •

Hypomagnesemia Hypovolemic shock Septic shock Generalized peritonitis Acute rumen acidosis Right-displaced abomasum/abomasal volvulus Hypokalemia Botulism Meningoencephalitis Polioencephalomalacia

TREATMENT Treatment of a nonambulatory cow evidently must focus on the primary cause of recumbency whenever it has been identified, but must also address secondary damage resulting from prolonged recumbency. The reader is referred to the corresponding chapter of this book for the treatment of primary cause of recumbency whenever it is known. Intensive supportive care is required for the treatment of secondary damage and prevention of further damage. The prognosis of a downer cow not only depends on the initial cause of the recumbency but to a large part also on the quality of the care provided during the recumbent period. Antiinflammatory Therapy Antiinflammatory therapy as part of pain management in ruminant production medicine has received increased attention over the past years because this is increasingly recognized as an essential aspect of animal welfare by veterinarians and owners alike.11 Although currently not much data are available to support the use of steroidal and nonsteroidal antiinflammatory drugs in downer cows, their use seems indicated not only to alleviate pain and discomfort of the sick, nonambulatory cow, but also to contain and control inflammation secondary to recumbency that is likely to exacerbate myopathy and neuropathy. Pain in cattle, as in other species, can occur as result of tissue damage, nerve damage, and inflammation, all factors considered to greatly contribute to downer-cow syndrome.12 Repeated doses of nonsteroidal antiinflammatory drugs (NSAIDs) may be required for adequate control of pain and inflammation, which may put the treated animal at increased risk for adverse gastrointestinal effects, such as abomasal ulceration.12 It is therefore advisable to instruct the patient owner to regularly check the produced feces for signs of melena. A single but high dose of dexamethasone (0.2 to 0.3 mg/kg IV) early in the recumbent period has been advocated by some clinicians, based on clinical experience, to control and contain inflammatory neuropathy resulting from trauma or pressure. Because of the abortive effect of the treatment, this therapy in pregnant cows must be discussed with the animal owner.

Fluid and Electrolyte Therapy Fluid and electrolyte therapy orally and if necessary parenterally is indicated in patients with inadequate water and feed intake. Multiple electrolytes can be added to the drinking water if the cow is drinking normally. The supplementation of minerals such as phosphates, magnesium, or potassium has been advocated, but they have been used without consistent success. Oral fluid therapy by drenching is an effective way to maintain hydration in an alert animal. For a recumbent cow, drenching should only be considered in alert cows with a good swallowing reflex. Because the pressure on visceral organs is increased with recumbency, the amount of fluid administered per treatment should not exceed 40 L to prevent the risk of reflux as a result of increased intraruminal pressure. Bedding and Clinical Care The most important aspect of treatment is to provide the most comfortable bedding possible and to roll the cow from side to side several times daily to minimize the extent of ischemic damage and para-analgesia that results from prolonged recumbency. With conscientious care and the provision of good bedding, palatable feed, and liberal quantities of water, most cows will attempt to stand with some difficulty and assistance within 24 hours, and most will stand unassisted and normally 1 or 2 days later. A sand or dirt pack is the ideal ground surface to facilitate standing when downer cows attempt to stand. If affected cows are left on a slippery ground surface, they will not make an effort to stand and will become progressively worse. Cows should be milked normally and the udder kept clean by washing with germicide soap before milking, and postmilking teat dips should be applied. Assisted Lifting to Aid Standing The clinician and farmer are commonly faced with the questions of whether or not to lift a recumbent cow that has not attempted to stand within a few hours after treatment for milk fever. The guiding principle should be the behavior of the cow. If the cow makes an effort to stand on her own or by some coaxing such as a gentle nudge in the ribs, she should be assisted to stand by ensuring a good nonslip ground surface, providing deep bedding, and lifting up on the tailhead when she attempts to stand. The cow should be rolled from side to side every few hours and encouraged to stand a few times daily. Several different kinds of cow-lifting devices have been used to assist downer cows to stand. Hip lifters, which fit and tighten over the tuber coxae, and body slings such as harnesses are designed to fit around the abdomen and thorax of the animal. These devices can assist a downer cow to stand if she makes some effort on her own. For those cows that make some effort to stand, the

1697

hip lifters or slings can be applied and the animal lifted to the standing position. If the animal bears weight on all four legs, she should be allowed to stand with the aid of the device for 20 to 30 minutes and then lowered down. This procedure can be repeated once or twice a day, provided the cow is able to support her own weight while standing. In most cases, such downer cows will stand on their own within a few days. While the cow is in the standing position, she can be milked, and other clinical examinations can be carried out. The hip lifters can result in traumatic injuries to the tissues surrounding the tuber coxae if not used judiciously. Cows carrying their own weight after being lifted must not under any circumstances be left unattended while hoisted with the hip lifter because they could lose strength and hang in the device, which could result in severe trauma. Animals that make no effort to stand and bear weight on their own must not be left suspended in the lifter for more than a few minutes but lowered immediately. If the hip lifters are not applied carefully, the animal may slip out of the device while it is being lifted, which commonly results in tissue injury around the tuber coxae; fractures of the coxae have even occurred. These injuries are often unnoticed clinically, but contribute to persistent recumbency. Lifting devices must be used carefully by experienced personnel. Body slings that fit around the abdomen and thorax of the animal are more suitable to lift down cows that will not readily bear weight after being hoisted, because they distribute the weight over several sites in contrast to the hip lifters, which concentrate the weight over the tuber coxae. However, the body slings are cumbersome to apply to a recumbent animal, and they require more time and experienced personnel to ensure proper application. When the slings are applied properly, they do appear to allow the lifted animal to stand comfortably for 30 minutes or more and promote recovery. Lifting cows that make no effort to stand on their own is usually unsuccessful. When lifted, they usually do not bear any significant weight. More recently, water flotation tanks have been used for the management of nonambulatory cows.3 Proposed devices consist of a watertight metal tub with inside dimensions of approximately 234 cm long, 109 cm wide, and 130 cm high. The system can be mobile, and although the use is labor intensive, it can give good results when selecting suitable patients judiciously.3 Depending on the system used the downer cow is either pulled into the tub on a mat and the ends of the tub closed to make a water-tight container with an open top like a bathtub or is fitted with a harness and lifted into the tub already filled with warm water. With the cow’s head held up by a halter, the tub is filled with water at 37° C to 38° C (100° F to 102°F) as quickly as

1698

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

possible. Cows in lateral recumbency will roll into sternal recumbency when 40 to 50 cm of water are in the tub and will usually attempt to stand when the tub is one-half to two-thirds full. Cows are allowed to stand in the water for 6 to 8 hours, in some cases up to 24 hours. If the water temperature falls below 35° C (95°F), more hot water is added. When the decision is made to remove the cow, the water is drained and the end of the tub opened, or the cow is hoisted out of the tub on a sling. A recent retrospective study including 51 recumbent patients of a veterinary teaching hospital treated with flotation tank reported a success rate of this therapy of 37%.3 The success rate could be higher if the selection of cases for flotation is more rigorous. Cows with ruptured tendons, fractures, luxated coxofemoral joints, septic polyarthritis, and other physical injuries of the musculoskeletal system are not good candidates for flotation. The most suitable case for flotation would appear to be the downer cow as a sequel to milk fever. Handling, Transportation, and Disposition of Nonambulatory Cattle There has been considerable controversy and disparity among veterinarians and livestock producers about the handling, transportation, and disposition of nonambulatory cattle. Economics has a major influence on decision making in these cases. There has been no common understanding of whether or not they are fit for transportation and which ones are fit for slaughter for salvage. When the owner and veterinarian are faced with a downer cow that is valuable and the cause of the recumbency is uncertain, the tendency is to either attempt to provide treatment for several days and assess the progress or consider slaughter for salvage. In the case of valuable breeding animals that are recumbent as a complication of milk fever, or a disease such as acute carbohydrate engorgement or peracute mastitis, supportive and specific therapies are commonly selected. In the case of downer cattle of commercial value, slaughter for salvage has been a common option. Cattle producers would like to obtain as much financial return as possible by slaughter for salvage. Cattle affected with complications of milk fever, traumatic injuries of the musculoskeletal system, and other diseases not associated with toxemia or septicemia are commonly submitted to slaughter for salvage. Transportation of these compromised animals has always been an animal welfare issue because of the difficulty of loading them humanely because of their size. The mere act of lifting, pulling, dragging, or by other means forcefully loading an animal weighing 500 to 800 kg onto a truck cannot be done without considerable pain and discomfort to the animal. However, beginning in the 1990s, worldwide concern emerged from the public about the handling and disposition of

nonambulatory animals, particularly downer cows, regardless of the cause of their recumbency. Government animal health regulatory agencies, livestock associations, and veterinary associations began drafting regulations on the care and handling of nonambulatory recumbent animals such as the downer cow. Downer-cow syndrome is an animal welfare issue, and the veterinarian should be proactive about the problem. Society is concerned about how downer animals are cared for and handled and the methods used for their disposition. If recovery does not occur within a few days, the prognosis is uncertain; the owner and veterinarian must decide whether to continue providing clinical care to the downer cow or whether the animal should be euthanized. Euthanasia The quality of care provided to a recumbent cow can easily become an issue of animal welfare, and humane euthanasia should always be considered, particularly in cases with poor prognosis or when the attending veterinarian can foresee that adequate supportive care cannot or will not be provided for whatever reason. Suggested “trigger points” for euthanasia suggested in the literature include the following:10 • Conditions with poor prognosis • Pain and suffering that is unresponsive to treatment • Anorexia over several days • Nonalert downer cows not responding to treatment in due time • Cows unable to maintain sternal recumbency • Owner unable or unwilling to provide adequate care • Complications such as pressure sore, mastitis, or other condition • Deterioration despite adequate patient care • Unresponsive to treatment for over 10 days TREATMENT AND CONTROL Treatment of primary cause as indicated Supportive care Move cow off concrete floor onto soft bedding (R-1) Oral fluid therapy in dehydrated or anorectic cows (R-1) Roll recumbent cow from one side to the other q4-8h (R-1) NSAIDs (at label dose with label treatment interval) (R-2) Dexamethasone (0.2 to 0.3 mg/kg IV as a single dose) (R-2) Hoist cows that make attempts to stand (R-2) Control Close monitoring of periparturient cows for signs of milk fever (R-1)

Immediate and adequate treatment of cows with milk fever (R-1) Provide comfortable calving area with soft bedding and nonslippery flooring (R-1) Avoid moving pregnant cows too late to calving area (R-2) Avoid moving fresh cows too early out of calving pen (R-2)

CONTROL The early detection and treatment of milk fever will reduce the incidence and severity of downer-cow syndrome. Under ideal conditions, cows should be treated during the first stage of milk fever before they become recumbent. Once recumbent, cows should be treated as soon as possible and not delayed for more than 1 hour. Cows with milk fever should be well bedded with liberal quantities of straw or moved to a soft-ground surface. Recumbent cows should be coaxed and assisted to stand if possible after treatment for milk fever. If they are unable to stand, they should be rolled from one side to the other every few hours if possible. It is usually difficult to get owners to comply with this recommendation, but frequent rolling from side to side is necessary to minimize the ischemic necrosis. Dairy cows should be placed in a comfortable, well bedded box stall before calving and should be left in that box stall until at least 48 hours after partition in the event that milk fever develops. FURTHER READING Cox VS. Nonsystemic causes of the downer cow syndrome. Vet Clin North Am Food Anim Pract. 1988;4:413-433. Grandin T. Welfare of cattle during slaughter and the prevention of non-ambulatory (downer) cattle. J Am Vet Med Assoc. 2001;219:1377-1382. Green AI, et al. Factors associated with occurrence and recovery of nonambulatroy dairy cows in the United States. J Dairy Sci. 2008;91:2275-2283. Poulton PJ. Musculo-Skeletal Examination and Diagnosis of the Downer Cow. Proc XXVIII World Buiatrics Congress. Cairns, Australia: 2014:212-218. Poulton PJ. Management of the Downer Cow. Proc XXVIII World Buiatrics Congress. Cairns, Australia: 2014:219-222.

REFERENCES

1. Stull CL, et al. J Am Vet Med Assoc. 2007;231: 227-234. 2. Green AL, et al. J Dairy Sci. 2008;91:2275-2283. 3. Burton AJ, et al. J Am Vet Med Assoc. 2009;234: 1177-1192. 4. NASS. Accessed July 12, 2015, at ; 2005. 5. Ménard L, Thompson A. Can Vet J. 2007;48: 487-4919. 6. Grünberg W. Vet Clin North Am Food Anim Pract. 2014;30:383-408. 7. Sattler N, Fecteau G. Vet Clin North Am Food Anim Pract. 2014;30:351-357. 8. Constable PD, et al. J Am Vet Med Assoc. 2013;242: 826-835.

Metabolic Diseases of Ruminants

VetBooks.ir

9. Poulton PJ. Proc. XXVIII World Buiatrics Congress. 2014:212-218. 10. Poulton PJ. Proc XXVIII World Buiatrics Congress. 2014: 219-222. 11. Thomsen PT, et al. Vet J. 2012;194:94-97. 12. Coetzee JF. Vet Clin North Am Food Anim Pract. 2013;29:11-28.

HYPOMAGNESEMIC TETANIES Tetany associated with a marked decrease in serum magnesium concentration is a common occurrence in ruminants. The syndrome associated with hypomagnesemia is relatively constant, irrespective of the cause, but the group of diseases in which it occurs has been divided into three groups: • hypomagnesemic tetany of calves, which appears to result specifically from a deficiency of magnesium in the diet transport tetany • a group of hypomagnesemias in ruminants characterized by lactation tetany, in which there may be a partial dietary deficiency of magnesium but in which nutritional or metabolic factors reduce the availability, or increase the body’s loss, of the element so that serum magnesium levels fall below a critical point. In general, the occurrence of hypomagnesemic tetany is related to three sets of circumstances. Most common is the occurrence in lactating cows turned out onto lush, grassdominant pasture in the spring after wintering in closed housing—the classic lactation or grass tetany of Holland. Wheat pasture poisoning may occur when cattle or sheep are grazed on young, green cereal crops. The third occurrence is in beef or dry dairy cattle running at pasture in the winter, usually when nutrition is inadequate and where no shelter is provided in changeable weather rather than in severe, prolonged cold. Less common forms occur in housed animals on poor feed. Hypomagnesemia of sheep, although less common, occurs in the same general groups of circumstances as the disease in cattle. A chronic hypomagnesemia, without manifestations of tetany, can be a cause of sub­optimal production efficiency and may predispose to hypocalcemia.

HYPOMAGNESEMIC TETANY (LACTATION TETANY, GRASS TETANY, GRASS STAGGERS, WHEAT PASTURE POISONING) SYNOPSIS Etiology The etiology is multifactorial, related to magnesium concentration in the diet and the presence of competing cations such as potassium and sodium that affect either herbage magnesium status or magnesium absorption.

Epidemiology Disease of all classes of ruminants, but reaches its highest incidence in older lactating cows exposed to bad weather or grazing green cereal crops or lush grass-dominant pasture. Clinical findings Incoordination, hyperesthesia and tetany, tonic–clonic muscular spasms and convulsions. High case fatality without treatment. Clinical pathology Serum, urine, vitreous humor, or cerebrospinal fluid (CSF) magnesium concentrations. Hypomagnesemia, and in some circumstances hypocalcemia. Necropsy findings None specific. Diagnostic confirmation Response to treatment, serum or urinary magnesium concentrations. Treatment Magnesium or combined calcium/ magnesium solutions administered IV or SC. Control Magnesium supplementation, but a palatable and practical delivery method is a problem. Magnesium applied to pastures. Avoidance of movement and food deprivation at risk periods.

ETIOLOGY

Magnesium is the major intracellular divalent cation and is an essential element in a large number of enzymatic activities in the body. For this reason, it might be expected that hypomagnesemia would be rare. However, because of the peculiarities of absorption of magnesium in the ruminant forestomaches, and the use of animal and pasture management systems that can lead to marginal magnesium uptake, ruminants are at risk of hypomagnesemia. Magnesium Homeostasis There is no feedback regulatory mechanism to control concentrations of magnesium in the body of ruminants. As a consequence, magnesium concentrations in blood and extracellular fluid are essentially determined by the balance between dietary intake of magnesium, loss in feces and milk, and the modulating effect of magnesium homeostasis by the kidney. Dietary Intake In normal circumstances, magnesium absorbed from the diet is sufficient to meet the requirements of the body, and excess amounts are excreted in the urine. Renal Excretion The kidney is the major organ of homeostasis and can act to conserve magnesium. Magnesium is freely filtered across the renal glomerulus and is reabsorbed within the renal tubules, the degree of reabsorption acting in homeostasis. The endogenous daily urinary loss is typically 3 mg/kg BW, equivalent to 1.8 g/day for a 600-kg cow. When the dietary intake of magnesium is decreased, blood and

1699

interstitial fluid magnesium concentrations fall; excretion of magnesium in the urine will cease when serum concentrations fall below 1.8 mg/dL. The renal threshold for magnesium excretion is partially under the control of parathyroid hormone, and increased plasma concentrations of parathyroid hormone will act to conserve magnesium. Magnesium Reserves There are large stores of magnesium in the body, especially in bone. These are available to the young calf, but mobilization rate decreases with age, and in the adult ruminant there is little mobilization in response to short-term deficits of magnesium. In ruminants, this control mechanism for magnesium can maintain adequate concentrations of magnesium in bodily fluids in most production circumstances, but it can fail where there is a high requirement for magnesium coupled with a decreased intake. This combination leads to hypomagnesemia, and hypomagnesemic tetany is a possible outcome. Lactation Increased requirement for magnesium is almost always associated with the loss of magnesium in the milk during lactation. Whereas the amount of magnesium in milk is not high (14 mg/kg BW), the loss of magnesium to milk in high-producing animals (4.2 g of magnesium in 30 L of milk) represents a significant proportion of the dietary intake of magnesium. As a consequence of this drain, most instances of hypomagnesemia occur in lactating animals around the period of peak milk production, although in some circumstances the demands of late pregnancy are the cause of the increased requirement. The decreased intake of magnesium can result from an absolute deficiency of magnesium in the diet or because the availability or absorption of magnesium from the diet is impaired. These factors determine the circumstances of occurrence of the disease and are the factors that can be manipulated for control. Factors Influencing Absorption of Magnesium In the adult ruminant, magnesium absorption occurs in the forestomach with little absorption in the abomasum and small intestine. Some absorption occurs in the large intestine, particularly in sheep; however, it cannot compensate for malabsorption in the forestomach. Magnesium is absorbed from the small intestine of calves, lambs, and kids, but this ability appears to be lost when these animals become ruminants. Na : K Ratio in Rumen Magnesium is transported across the epithelium of the forestomaches by an active sodium-linked ATPase-dependent transport system. Absorption, and the serum magnesium concentration, is influenced by the

1700

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Na : K ratio in the rumen, which is determined by the dietary and salivary concentrations of sodium and potassium. Absorption of magnesium increases with an increasing Na : K ratio to plateau at a ratio of 5 : 1. Absorption is significantly impaired if the Na : K ratio is less than 3 : 1. Young, rapidly growing grass that is low in sodium and high in potassium can result in sodium deficiency in grazing ruminants and can significantly depress the Na : K ratio in the rumen fluid, causing impaired magnesium absorption. Depression is observed at dietary potassium concentrations of greater than 22 g/kg dry matter. Saliva normally has a high Na : K ratio, but where there is a deficit of sodium in the diet, a proportion of sodium in saliva may be replaced with potassium under the influence of aldosterone, which further negatively influences the uptake of magnesium. Approximately 40% of the total magnesium available in extracellular fluid is secreted daily in saliva, and 20% of this is reabsorbed in the forestomach. When animals are on tetany-prone grass, forestomach absorption is impaired, which accounts for the susceptibility of ruminants to hypomagnesemia compared with monogastric animals. Other Factors Influencing Absorption Young grass fertilized with nitrogenous fertilizers has an increased crude protein content, which is readily fermentable and leads to increased ammonia concentrations. A sudden rise in ruminal concentrations of ammonia impairs magnesium absorption in the rumen. The uptake of magnesium is also influenced by the carbohydrate content of the diet; magnesium absorption is improved with increasing amounts of readily degradable carbohydrates. The mechanism of this action is not known, but low concentrations of readily degradable carbohydrate in tetany-prone pastures in combination with high concentrations of protein may be important to the occurrence of the syndrome. Volatile fatty acids provide the energy for the active transport of magnesium across the rumen wall and increase magnesium absorption. Ruminal pH is thought to affect absorption efficiency by influencing magnesium solubility, which decreases markedly as ruminal pH increases above 6.5. Magnesium binders, such as fats, can form insoluble magnesium salts. Other dietary substances have been proposed to influence the absorption of magnesium, including calcium and phosphorus, organic acids such as citric acid and transaconitate, fatty acids, and aluminum, but the significance of their role is controversial. Magnesium in Pastures and Tetany Hazard The dietary intake of magnesium in grazing animals is directly related to the magnesium

concentration in pastures, but other elements in pastures also influence magnesium absorption by the ruminant, as detailed earlier. Required Magnesium Concentrations Hypomagnesemia can result from the ingestion of pastures that have insufficient magnesium to meet dietary requirements. The estimated magnesium concentration in pasture required to meet the dietary requirement for pregnant or lactating cattle varies from 1.0 to 1.3 g/kg dry matter (DM) for pregnant cattle, depending on the stage of pregnancy, and from 1.8 to 2.2 g/kg DM for lactating cattle, with both estimates assuming minimal interference of absorption by other elements in the pasture. The recommended minimal “safe” concentration of magnesium in pastures is 2 g/ kg DM for lactating and pregnant cattle, with a preference for a concentration of 2.5 g/kg DM. Magnesium Availability in Pastures and Hazard Hypomagnesemia can also occur in animals grazing pastures with adequate concentrations of magnesium but that contain high concentrations of potassium and nitrogen, which, as detailed earlier, impair absorption of magnesium in the rumen. Pastures with concentrations of potassium of greater than 30 g K/kg DM and nitrogen greater than 40 g N/kg dry matter are considered hazardous. An alternate method for estimating the potential hazard of a pasture is to calculate the K/(Ca + Mg) ratio using milliequivalent (mEq) values for this estimate. Pastures with ratios above 2.2 are considered a risk. Winter Hypomagnesemia The occurrence of hypomagnesemia is not restricted to cattle grazing lush pastures; it can also occur during winter. In housed lactating dairy cattle being fed conserved feeds, hypomagnesemia probably has the same genesis as that in grazing cattle, being associated with a high lactational drain of magnesium in combination with the feeding of conserved feeds prepared from pastures with marginal magnesium concentrations. Hypomagnesemia also occurs in cattle outwintered on poor-quality feed. Hypomagnesemia and Hypocalcemia In some outbreaks of hypomagnesemic tetany, there is also hypocalcemia, and although it is of less severe degree than in parturient paresis, there is increasing evidence that the actual onset of clinical tetany may be associated with a rapid fall in serum calcium levels superimposed on a preexisting hypomagnesemia. This is particularly true for wheat pasture poisoning but can also apply to outbreaks with different predisposing factors. Chronic hypomagnesemia can have a profound effect on calcium homeostasis. Hypomagnesemia reduces the production

and secretion of parathyroid hormone, reduces hydroxylation of vitamin D in the liver, and also causes target-organ insensitivity to the physiologic effects of parathyroid hormone and 1,25-dihydroxyvitamin D3. Chronic subclinical hypomagnesemia can increase susceptibility to milk fever and can predispose to episodes of milk fever and downer cows in lactating dairy cows during the period of peak lactation. Summary of Etiology In summary, it appears that a number of factors are capable of causing hypomagnesemia in ruminants and that under particular circumstances one or other of them may be of major importance. In lactation tetany of cows and ewes turned out onto lush pasture in the spring, a primary dietary deficiency of magnesium or the presence of high relative concentrations of potassium and nitrogen in the diet reduces the absorption of magnesium and possibly calcium. In wheat (cereal) pasture poisoning, the ingestion of abnormally large amounts of potassium and low levels of calcium in the diet leads to hypomagnesemia and also hypocalcemia. Hypomagnesemic tetany in cattle wintered at pasture and exposed to inclement weather is associated with low magnesium intake and inadequate caloric intake, and possibly to the resultant hyperactivity of the thyroid gland. Although the suggestions as to the most important etiologic factors in each set of circumstances in which lactation tetany occurs may be valid, undoubtedly combinations of these and other factors have etiologic significance in individual outbreaks of the disease. The worst combination of causative factors, and the most common circumstances in which the disease occurs, is inadequate energy intake with a low dietary content of magnesium (grass pasture) in recently calved cows during a spell of cold, wet, and especially windy weather. One other important factor is the variation between individual animals in susceptibility to hypomagnesemia and to the clinical disease. These variations are quite marked in cattle, and in intensively managed, high-producing herds it is probably worthwhile to identify susceptible animals and give them special treatment.

EPIDEMIOLOGY Occurrence and Risk Factors for Lactation Tetany Lactation tetany in dairy and beef cattle turned out to graze on lush, grass-dominant pasture after winter housing is common in northern Europe, the United Kingdom, and the northern parts of North America. Grass tetany also occurs in Australia and New Zealand, where the cows are not housed in winter but have access to a phenomenal flush

Metabolic Diseases of Ruminants

VetBooks.ir

of pasture growth in the spring. This also commonly occurs in beef cattle in all countries. With housed cattle, or cattle fed conserved feed during the winter, most cases occur during the first 2 weeks after the cattle are turned out to spring pasture. Pasture that has been heavily top dressed with fertilizers rich in nitrogen and potash is potentially the most dangerous. The disease may also occur on this type of pasture even when the cattle have wintered on pasture in temperate regions. In regions where there is an autumn flush of pasture, a high incidence of hypomagnesemic tetany may occur in the autumn or early winter. Cattle in the first 2 months of lactation and 4 to 7 years of age are most susceptible, which probably reflects an increased risk because of a higher loss of magnesium in milk. Friesian cows have lower magnesium concentrations than Jerseys grazed under the same conditions. In the northern parts of the United States, outbreaks commonly occur during periods of low barometric pressure when the ambient temperature ranges between 7° C (45° F) and 16° C (60° F) and soil temperatures are below 7° C (45° F). Outbreaks may be precipitated by inclement weather. In beef cattle there is commonly a history of poor nutrition and falling body condition in the past few weeks as a result of diminishing hay supplies. Occurrence and Risk Factors for Wheat (Cereal) Pasture Poisoning Wheat pasture poisoning is a misnomer because it can occur with grazing of any small-grain cereal pasture. It has been recorded in many countries, but it is most prevalent where young cereal crops are utilized for winter grazing. The southwestern United States has experienced heavy losses of cattle caused by this disease. This pasture can induce hypomagnesemia in pregnant and lactating cattle and sheep. The risk is with young, rapidly growing pasture, either in the spring or in the autumn and winter with pastures planted in late summer. The pasture is usually dangerous for only a few weeks, but heavy losses may occur in all classes of sheep and cattle. Bos taurus breeds are more susceptible to the development of hypomagnesemia than Bos indicus. Occurrence and Risk Factors for Winter Hypomagnesemia Hypomagnesemic tetany in cattle wintered in the open causes some losses in the United Kingdom, New Zealand, southern Australia, and the east-central states and Pacific slope of the United States. It occurs in cattle grazed on pasture in the winter with minimal supplemental hay and in cattle grazed on aftermath crops and corn stover. The disease occurs in regions with temperate climates, and risk is increased by exposure to bad

1701

weather, which is exacerbated by absence of trees or other shelter in fields and by failure to supply supplementary feed during these cold spells. The disease does not seem to occur in cattle kept outside in prolonged winters where environmental temperature is consistently very low and there is adequate feed. Hypomagnesemia, commonly presenting as chronic hypomagnesemia and sudden death, has been recognized as occurring in housed cattle in the winter in Europe for many years and recently has also been reported in the United States.

differences, and grasses with a high ratio of potassium to calcium and magnesium (e.g., Dactylis glomerata, Lolium perenne, Phalaris arundinacea) are more likely to cause grass tetany than those with low ratios (e.g., Bromus inermis, Poa pratensis, Agrostis spp.). On soil types where the disease is common, cool-season grass pastures top dressed with nitrogenous fertilizers are dangerous, and their toxicity may be increased by the application of potash. Warm-season grasses do not have the same risk, and grass tetany is not a problem in cattle grazing tropical grasses.

Morbidity and Mortality In all of these forms of the disease, the morbidity rate is highly variable, reaching as high as 12% in individual herds and up to 2% in particular areas. The incidence varies from year to year depending largely on climatic conditions and management practices, and the disease is often limited in its occurrence to particular farms and even to individual fields. Although an effective treatment is available, the case-fatality rate is high because of the short course. Because animals die before they are observed to be ill, there are not accurate figures on case fatality, but it is probably of the order of 30% in dairy cattle and considerably higher in beef cattle. There have been few epidemiologic studies specifically addressing the importance of the syndrome. In Finland, a lactational incidence varying between 0.1% and 0.3% has been recorded, with an increase in parity to at least six for lactation tetany occurring on pasture but not for indoor tetany. No association with other diseases was found other than for milk fever. In Northern Ireland, approximately 10% of dairy cows and 30% of beef cows have subnormal or deficient blood magnesium concentrations during the grazing season, and hypomagnesemia is considered the cause of 20% of the sudden-death mortality in beef cattle. Surveys of beef cattle owners of the relative importance of different diseases invariably rate hypomagnesemia high in importance.

Cereal Pastures The greater tendency of cereal grazing to cause hypomagnesemia is related to a high content of potassium and a low content of magnesium. The tetany hazard, in order of decreasing hazard, is wheat, oats, barley, rye.

Pasture Risk Factors In most areas of the world, there is a strong association between risk for hypomagnesemia and systems of pasture improvement and pasture fertilization to increase forage yield. There are a number of influences on the concentration of magnesium and other elements in pasture. Pasture Species Hypomagnesemia is a problem on grassdominant pastures. Concentrations of calcium and magnesium are higher in legumes and forbs than in grasses. Within the grasses, different genotypes of the same species can differ markedly in calcium and magnesium concentrations, and most coolseason grasses have the potential to produce hypomagnesemia. However, there are some

Season High concentrations of potassium and nitrogen and low concentrations of sodium and soluble carbohydrates occur in pastures during the early growing season and during rapid growth following cold, wet periods. Pasture magnesium concentrations may not be depressed, but the K/(Ca + Mg) ratio is increased. Fertilization Application of potash and nitrogenous fertilizers to pastures will decrease the concentration of calcium and magnesium in plants and will also increase the concentration of potassium and nitrogen. There is some evidence that nitrate sources of nitrogen depress magnesium less than ammonium sources of nitrogen. Soil Type The availability of magnesium to the plant is influenced by soil type, and some deficiencies in plant magnesium can be corrected by soil fertilization with magnesium. There is no strong association with any one soil type, but high potassium concentrations are consistently associated with increased risk for tetany. Highly leached, acid, sandy soils are particularly magnesium deficient and the most likely to respond to liming and magnesium fertilization. In very acidic soils, high aluminum concentrations may depress magnesium uptake by plants. A local knowledge of soil type and its influence on magnesium, potassium, calcium, and nitrogen uptake by pastures can allow the judicious selection or avoidance of the use of pastures for at-risk groups during periods of risk for hypomagnesemia. Animal and Management Risk Factors Dry Matter Intake The dry matter and energy intake of ruminants can influence susceptibility to

1702

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

hypomagnesemia. A reduction in dry matter intake must reduce the magnesium intake; in situations where hypomagnesemia is already present, a further depression of serum magnesium levels can be anticipated when complete or partial starvation occurs. An insufficient intake of fiber in the winter months can precipitate hypomagnesemia in pastured cows and ewes, and lipolysis is accompanied by a fall in serum magnesium. Period of Food Deprivation Many outbreaks of hypomagnesemia are preceded by an episode of stress or temporary starvation. Whether chronic hypomagnesemia preexists or not, a period of starvation in lactating cows and ewes is sufficient to produce a marked hypomagnesemia, and the fall may be sufficiently great to cause clinical tetany. A period of bad weather, yarding, transport, or movement to new pastures or the introduction to unpalatable pastures may provide such a period of partial starvation. Alimentary Sojourn Diarrhea is commonly associated with lactation tetany on spring pasture and, by decreasing the alimentary sojourn, may also reduce magnesium absorption. Climate A close association between climatic conditions and serum magnesium levels has also been observed. Reduced levels occur in adult cattle and sheep exposed to cold, wet, windy weather with little sunshine and no access to shelter or supplementary feed. Supplementary feeding appears to reduce the effect of inclement weather on serum magnesium levels, and it is possible that failure to eat, or depression of appetite, and a negative energy balance during bad weather may be a basic contributing cause to hypomagnesemia in these circumstances. Animal Movement Epinephrine release will result in a precipitous fall in serum magnesium, and this may explain the common observation that clinical cases are often precipitated by excitement or movement of the herd. Intensive Dairies Intensive dairies that apply effluent on a limited land base can build soil potassium to high concentrations. Silage from these grounds can have a high risk for inducing hypomagnesemia. Hypomagnesemia in Sheep Hypomagnesemia occurs in sheep, particularly in Australia and the United Kingdom. The disease is not common, but it appears to be increasingly associated with pasture improvement practices, and it can cause heavy losses in individual flocks. It is more

common in ewes bred for milk and lamb production. In outbreaks, ewes with twins are more liable to develop clinical disease than those with singles, and the main occurrence is in ewes 1 to 4 weeks after lambing, with cases up to 8 weeks after lambing. Disease is often precipitated by a management procedure involving movement and temporary food deprivation, and cases will occur within the first 24 hours following this and for a few days afterward. As in cattle, disease occurs when ewes are placed on lush grass pastures, but it is especially common where ewes in early lactation are placed on young cereal pastures. Losses usually cease when the flock is moved onto rough, unimproved pasture. Cases also occur in sheep that are exposed to inclement weather when on low nutritive intake. Simultaneous hypomagnesemia and ketosis can occur in ewes after lambing if they are exposed to low feed availability. These cases do not respond well to treatment. Hypomagnesemia in ewes is predisposed by prior pregnancy toxemia in the flock.

PATHOGENESIS

Most evidence points to hypomagnesemia as the cause of the tetanic signs observed, but the concurrent hypocalcemia may have a contributory effect and in many instances may even be the dominant factor. Most clinical cases of the disease have serum magnesium concentrations below 1 mg/dL (0.4 mmol/L) compared with the reference range in cattle of 1.7 to 3.0 mg/dL (0.7 to 1.2 mmol/L), and there is a striking relationship between the incidence of the clinical disease and the occurrence of a seasonal hypomagnesemia. The reduction in serum concentrations of magnesium is concurrent with a marked fall in the excretion of magnesium in the urine. In affected herds and flocks, many clinically normal cows and sheep have low serum magnesium concentrations. In some of these circumstances a concurrent hypocalcemia may be the precipitating cause. Magnesium has many influences on impulse transmission in the neuromuscular system, including effects on the release of acetylcholine, on the sensitivity of the motor end plate, on the threshold of the muscle membrane, and on activation of the cholinesterase system. These offer an attractive hypothesis for the muscular irritability seen with the disease. However, it has also been established that magnesium concentrations in the cerebrospinal fluid (CSF) are more predictive of clinical disease than those in serum, which would indicate that alterations in central nervous system (CNS) function are more important than alterations in peripheral nerve function. It is also evident that CSF concentrations of magnesium in hypomagnesemic animals rise significantly after treatment with a magnesium salt. The need for this to happen would explain the

delay of about 30 minutes after an IV injection before recovery occurs, because CSF volume turns over at approximately 1% per minute.

CLINICAL FINDINGS

For convenience, lactation tetany is described in acute, subacute, and chronic forms. Acute Lactation Tetany In acute lactation tetany, the animal may be grazing at the time and suddenly cease to graze, adopt a posture of unusual alertness, and appear uncomfortable; twitching of the muscles and ears is also evident. There is severe hyperesthesia, and slight disturbances precipitate attacks of continuous bellowing, frenzied galloping, and occasionally aggression. The gait becomes staggering, and the animal falls, with obvious tetany of the limbs, which is rapidly followed by clonic convulsions lasting for about a minute. During the convulsive episodes the following characteristics are common: • Opisthotonos • Nystagmus • Champing of the jaws • Frothing at the mouth • Pricking of the ears • Retraction of the eyelids Between episodes, the animal lies quietly, but a sudden noise or touch may precipitate another attack. The temperature rises to 40.0 to 40.5° C (104 to 105° F) after severe muscle exertion; the pulse and respiratory rates are also high. The absolute intensity of the heart sounds is increased so that they can be heard some distance away from the cow. Death usually occurs within 5 to 60 minutes, and the mortality rate is high because many die before treatment can be provided. The response to treatment is generally good if the animal is treated early. Subacute Lactation Tetany In subacute lactation tetany, the onset is more gradual. Over a period of 3 to 4 days, there is slight inappetence, wildness of the facial expression, and exaggerated limb movements. The cow often resists being driven and throws her head about as though expecting a blow. Spasmodic urination and frequent defecation are characteristic. The appetite and milk yield are diminished, and ruminal movements decrease. Muscle tremor and mild tetany of the hindlegs and tail with an unsteady, straddling gait may be accompanied by retraction of the head and trismus. Sudden movement or noise, the application of restraint, or the insertion of a needle may precipitate a violent convulsion. Animals with this form of the disease may recover spontaneously within a few days or progress to a stage of recumbency with a similar but rather milder syndrome than in the acute form. Treatment is usually effective, but there is a marked tendency to relapse.

Metabolic Diseases of Ruminants

VetBooks.ir

Chronic Hypomagnesemia Many animals in affected herds have low serum magnesium levels but do not show clinical signs. There may be sudden death. A few animals exhibit a rather vague syndrome that includes dullness, unthriftiness, and indifferent appetite and may subsequently develop one of the more obvious syndromes. In lactating cows, this may be the development of paresis and a milk-fever-like syndrome that is poorly responsive to calcium treatment. Depressed milk production has also been attributed to chronic hypomagnesemia in dairy herds in New Zealand. The chronic type may also occur in animals that recover from the subacute form of the disease. Parturient Paresis With Hypomagnesemia This syndrome is described in the discussion of parturient paresis and consists of paresis and circulatory collapse in an adult cow that has calved within the preceding 48 hours but in which dullness and flaccidity are replaced by hyperesthesia and tetany.

CLINICAL PATHOLOGY

Serum or urinary magnesium concentrations can be used for clinical cases. Where an animal is dead and hypomagnesemia is suspect, a presumptive diagnosis can be made from samples taken from other at-risk animals in the group or from the vitreous humor of the dead animal. An acute-phase inflammatory response with leukocytosis and increased numbers of neutrophils and monocytes has been recorded in ruminants and laboratory animals fed magnesiumdeficient diets. Serum Magnesium Concentrations Normal serum magnesium concentrations are 1.7 to 3.0 mg/dL (0.70 to 1.23 mmol/L). These levels in cattle are often reduced in seasonal subclinical hypomagnesemia to between 1 and 2 mg/dL (0.41 and 0.82 mmol/L), but risk for tetany is not present until the level falls to below 1.2 mg/ dL (0.49 mmol/L). The average concentration at which signs occur is approximately 0.5 mg/dL (0.21 mmol/L), and in sheep it is suggested that clinical tetany does not occur until the serum magnesium concentration is below 0.5 mg/dL (0.21 mmol/L). Serum magnesium concentration in some animals may fall to as low as 0.4 mg/dL (0.16 mmol/L) without clinical illness. This may be a result of individual animal variation in the degree of ionization of the serum magnesium and in the difference between serum and CSF concentrations. A transitory elevation of serum magnesium concentration occurs after violent muscular exercise in cattle with clinical signs of hypomagnesemia. Total serum calcium concentrations are often reduced to 5 to 8 mg/dL (1.25 to 2.00 mmol/L), and this may have an

1703

important bearing on the development of clinical signs. Serum inorganic phosphate concentrations may or may not be low. In wheat pasture poisoning of cattle there is hypomagnesemia, hypocalcemia, and hyperkalemia. In acute tetany, serum potassium concentrations are usually dangerously high and may contribute to the high death rate. Magnesium Concentrations in Cerebrospinal Fluid Magnesium concentrations in CSF can be used as a diagnostic procedure, but CSF is not easily or safely collected in tetany cases. CSF collected up to 12 hours after death can be used diagnostically. Magnesium concentrations in CSF of 1.25 mg/dL (0.51 mmol/L) were found in tetanic cows with hypomagnesemia (serum magnesium concentrations of 0.54 ± 0.41 mg/ dL; 0.22 ± 0.17 mmol/L). In clinically normal cows with hypomagnesemia, comparable concentrations in CSF were 1.84 mg/dL (0.74 mmol/L) and 0.4 mg/dL (0.16 mmol/L) in serum. In normal animals CSF concentrations are the same as in plasma, that is, 2.0 mg/ dL (0.82 mmol/L) and up. The magnesium content of ventricular CSF may be quite different from that of lumbar CSF. Ventricular CSF is also more responsive to changes in serum magnesium concentrations and is preferred for diagnosis at necropsy. Vitreous Humor Magnesium concentrations in vitreous humor (but not aqueous humor) can be measured because vitreous humor magnesium concentration remains stable for a longer period of time than magnesium concentrations in serum or CSF. In general, vitreous humor magnesium concentrations less than 0.55 mmol/L for cattle and less than 0.65 mmol/L for sheep up to 48 hours after death indicate the presence of hypomagnesemia, particularly at ambient temperatures of 4° C or 20° C.1 Vitreous humor is viscous and must be collected using a 14-gauge (preferably) or 16-gauge needle attached to a syringe. With the deceased animal placed in sternal recumbency, the needle is introduced vertically from a position caudal to the dorsal limbus of the eye parallel and caudal to the lens before aspiration (Fig. 17-5). The needle position should be altered to facilitate aspiration.1 The aspirated sample should be placed in a plain tube and centrifuged, and the supernatant should be submitted for determination of the magnesium concentration. Aqueous humor should not be collected for analysis because it is readily contaminated by degenerating iris tissue and evaporation of free water across the cornea. Urine Magnesium Concentrations The occurrence of low urine magnesium levels is good presumptive evidence of

Fig. 17-5  Vitreous humor is sampled from a recently deceased animal by inserting a 14-gauge needle perpendicular and caudal to the limbus. The needle tip can be observed through the pupil. Aqueous humor is obtained by inserting a 21-gauge needle horizontally rostral to the limbus and into the anterior chamber. Vitreous humor is required to evaluate magnesium concentrations. (Reproduced, with permission, from Edwards G, Foster A, and Livesey C. In Practice 2009; 31:22-25.)

hypomagnesemia; however, it is not the most sensitive test. Normalization of urine magnesium to simultaneously determine urine creatinine concentration will adjust urine magnesium concentration for animal-toanimal variability in urine concentration.2 Further adjustment by calculating the fraction clearance of magnesium (requiring simultaneous determination of plasma/ serum magnesium and creatinine concentrations) has not been shown to provide additional information beyond that provided by the concentration of urinary magnesium to creatinine alone. Herd Diagnosis The kidney is the major organ of homeostasis, and it has been argued that analysis of urine magnesium status is a more accurate method of assessing herd magnesium status than serum magnesium concentrations. The magnesium status of a herd, and the need to supplement the diet to prevent lactation tetany, can be established from the following values: • serum magnesium concentrations • urine magnesium concentrations • urinary magnesium fractional clearance2 • creatinine-corrected urinary magnesium concentrations Laboratory charges for urinary magnesium fractional clearance ratios are expensive. The determination of the creatinine-corrected urinary magnesium concentration from 10 cows in a herd has been found to be a more sensitive indicator of magnesium status of the herd than estimates from serum, and it is a better predictor of response to supplementation. Values of less than 1.0 mmol/L indicate that a positive response to supplementation is likely. Urine

1704

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

magnesium concentrations below 1.0 mg/ dL (0.4 mmol/L) indicate a danger for tetany.

adipate and 5% calcium gluconate at a dose rate of 500 mL is also used.

NECROPSY FINDINGS

Magnesium Therapy When magnesium solutions are used, 200 to 300mL of a 20% solution of magnesium sulfate may be injected IV; this is followed by a very rapid rise in serum magnesium concentration, which returns to preinjection levels within 3 to 6 hours. A much slower rise and fall occurs after SC injection, and for optimum results the SC injection of 200 mL of a 50% solution of magnesium sulfate has been recommended. A rise in serum magnesium of 0.5 mg/dL (0.21 mmol/L) occurs within a few minutes, and subsequent serum concentrations do not go above 5 mg/dL (2.06 mmol/L). In cases where serum magnesium concentrations are low because of seasonal hypomagnesemia, the injection of magnesium salts is followed by a rise and then a return to the subnormal preinjection levels. The IV injection of magnesium salts is not without danger. It may induce cardiac dysrhythmia, or medullary depression may be severe enough to cause respiratory failure. If signs of respiratory distress or excessive slowing or increase in heart rate are noticed, the injection should be stopped immediately and, if necessary, a calcium solution injected. The substitution of magnesium lactate for magnesium sulfate has been recommended to provide a more prolonged elevation of serum magnesium levels. A dilute solution (3.3%) causes minimal tissue injury and can be administered IV or SC. Magnesium gluconate has also been used as a 15% solution at dose rates of 200 to 400 mL. High serum magnesium concentrations are obtained more slowly and are maintained longer with magnesium gluconate than with magnesium sulfate. The feeding of magnesium-rich supplements, as described in the following section on control, is recommended after parenteral treatment.

There are no specific necropsy findings. Extravasations of blood may be observed in subcutaneous tissues and under the pericardium, endocardium, pleura, peritoneum, and intestinal mucosa. Agonal emphysema may also be present. The magnesium content of the bovine vitreous humor is considered to be an accurate estimate of magnesium status for 72 hours after death, provided the environmental temperature does not exceed 23° C (73° F) and there is not growth of bacterial contamination after sampling, which can result in a false low magnesium concentration. The addition of a small amount of 4% formaldehyde (3% of the vitreous humor volume) will allow accurate analysis for periods up to 72 hours after sampling. Concentrations in the aqueous humor are not stable after death.

DIFFERENTIAL DIAGNOSIS Cattle • Acute lead poisoning • Rabies • Nervous ketosis • Bovine spongiform encephalopathy Sheep • Hypocalcemia • Phalaris poisoning • “Stagger” syndromes

TREATMENT IV administration of preparations containing magnesium or, more commonly, magnesium and calcium are used. The efficiency of the various treatments appears to vary from area to area, and even within areas under different conditions of management and climate. Response rates and recovery rates are much higher in cases treated early in the clinical course. IV chloral hydrate may be administered to reduce the severity of convulsions during treatment with magnesium. Case fatality, even with therapy, can be high, especially in advanced cases. Combined Calcium/Magnesium Therapy The safest general recommendation is to use a combined calcium–magnesium pre­ paration (e.g., 500 mL of a solution containing 25% calcium borogluconate and 5% magnesium hypophosphite for cattle, 50 mL for sheep and goats) IV followed by an SC injection of a concentrated solution of a magnesium salt. The details and risks of administration of the type of solution are described in the section on parturient paresis. A combination of 12% magnesium

Provision for Further Cases The predisposing factors that lead to a case of hypomagnesemia apply to the herd as a whole, and it is probable that further clinical cases will occur before the effects of corrective strategies are observed. In extensive range situations, it is advisable to instruct the owner on how to treat cases because a delay in treatment can markedly increase the rate of treatment failures. SC treatment is within the realm of most, but successful therapy is also recorded by the rectal infusion of 30 g of magnesium chloride in a 100-mL solution; serum concentrations of magnesium return to normal levels within 10 minutes of administration.

CONTROL

Where possible, animals at high risk should be moved to low-risk pastures during the grass tetany season. High-risk pastures can

be grazed by low-risk animals, steers, or yearling heifers, for example, during this period. The occurrence of hypomagnesemia can be corrected by the provision of adequate or increased amounts of magnesium in the diet. A requirement as high as 3.0 g/kg DM diet may be required for lactating cows on spring pasture. The problem is in determining an adequate delivery system, and this will vary according to the management system. Thus blocked minerals containing magnesium or foliar dressing of magnesium may be adequate delivery systems where there is a high stocking density of cattle, but they are totally inadequate or economically unfeasible on range with one cow per 20 acres. Magnesium oxide is commonly used for supplementation, but other magnesium salts can be used, and they have an approximate equivalent availability. The biological availability of magnesium from magnesium carbonate, magnesium oxide, and magnesium sulfate for sheep is influenced by particle size, but it has been determined as 43.8%, 50.9%, and 57.6%, respectively. Feeding of Magnesium Supplements The preventive measure that is now universally adopted is the feeding of magnesium supplements to cows during the danger period. The feeding of magnesite (containing not less than 87% magnesium oxide), or other sources of magnesium oxide, prevents the seasonal fall in serum magnesium concentrations. Daily administration by drenching, or in the feed, of at least 60 g of magnesium oxide per day is recommended to prevent the disease. This is not always completely effective, and in some circumstances large doses may be necessary. Daily feeding of 120 g is safe and effective, but 180 g daily may cause diarrhea. The dose for sheep and goats is 7 g daily or 14 g every second day. Magnesium phosphate (53 g/d) is also a safe and effective way of ensuring a good intake of magnesium. The protection afforded develops within several days of commencing administration and terminates abruptly after administration ceases. Problems With Palatability The problem with magnesium supplements is getting the stock to eat the required amount because they are unpalatable. This can be partially countered by mixing the supplement with molasses in equal parts and allowing free access to the mixture or feeding it in ball feeders, but uniform intake by all animals does not occur, and at-risk animals may still develop hypomagnesemic tetany. Similarly, magnesium blocks may have limited efficacy in preventing hypomagnesemia. Salt blocks can help repair the sodium deficiency associated with young spring grasses and improve the Na : K ratio in the rumen. If they also contain Mg they can be

Metabolic Diseases of Ruminants

VetBooks.ir

an aid in prevention, but usually, by themselves, they do not guarantee freedom from risk for tetany. Spraying on Hay One method of attempting to ensure an adequate intake of magnesium is to spray it on hay and to feed this hay as a supplement during periods of grass tetany risk. The common practice is as follows: 1. Mix magnesite with molasses. 2. Dilute mixture with water. 3. Spray mixture onto hay in the windrows when it is being made. 4. Inject mixture into the bales before feeding or spray onto the hay at feeding. 5. Tip bale of hay so that the cut side is uppermost and pour the mixture evenly over the entire surface area. 6. Determine the level of application by the amount of hay intended to be fed. Depending on local circumstances, this method may or may not be effective because cattle and sheep will frequently not eat hay when on spring pasture unless they are confined for that purpose. Pellets Magnesium-rich pellets suggest themselves as a means of supplementation when the additional cost can be borne. Palatability is again a problem, and care needs to be taken to include palatable material in the pellets; alternatively, they may be mixed with other grain or molasses for feeding. Calves should be restricted from access because magnesium oxide at high levels of intake (2% and 4% of the ration) is toxic to calves and causes diarrhea with much mucus in the feces. In some high-risk situations it may be advisable to provide magnesium in several forms to ensure adequate intake. Routine Daily Drenching A once-daily oral administration of magnesium oxide or magnesium chloride to lactating dairy cows (to provide 10 g magnesium per cow), administered with a drenching gun just before the cows leave the milking parlor, is used in New Zealand to ensure adequate supplemental magnesium during periods of high risk. The cows become used to the procedure (and the farmers adept at carrying it out), and it causes minimal disruption of management. Heavy Magnesium “Bullets” The use of heavy “bullets” of magnesium to prevent hypomagnesemia has been effective in laboratory trials, and they are available commercially in some countries. The objective is to place a heavy bullet of magnesium in the reticulum, from which it constantly liberates small amounts of magnesium— about 1 g/d. This objective is achieved, and the occurrence of the clinical disease is

usually greatly reduced but not eliminated. In dangerous situations, it is customary to administer up to four bullets at a time. As with all bullets, there is a proportion lost by regurgitation and by passage through the gut. A special sheep-sized bullet is used in ewes, with similar results. Top Dressing of Pasture Top dressing of pasture, together with magnesium-rich fertilizers, raises the level of magnesium in the pasture and decreases the susceptibility of cattle to hypomag­ nesemia. For top dressing, calcined magnesite (1125 kg/ha) or magnesic limestone (5600 kg/ha) are satisfactory, with the former resulting in a greater increase in pasture magnesium. Other magnesium-containing fertilizers can be used depending on cost. The duration of the improved magnesium status varies with the type of soil: it is greatest on light sandy loams, on which a dressing of 560 kg/ ha of calcined magnesium can provide protection for 3 years. On heavy soils protection for only 1 year is to be expected. To avoid unnecessary expense, it may be possible to top dress one field with the magnesium fertilizer and keep this field in reserve for spring grazing. Fertilization with magnesium is expensive, and the response of pastures varies markedly with the soil type. It is advisable to seek agronomic advice. Foliar Dusting and Spraying The magnesium content of pastures can be raised much more quickly by spraying with a 2% solution of magnesium sulfate at fortnightly intervals or by application of very finely ground magnesium oxide to the pasture (30 kg/ha) before grazing commences. The technique is referred to as foliar dusting or spraying and has the advantage over feed supplementation that the intake is standard. It is very effective in cattle in maintaining serum magnesium concentrations and preventing the occurrence of the clinical disease. Dusting with 20 to 50 kg MgO/ha can provide protection for up to 3 weeks, but the duration is adversely influenced by wind and rain. A MgO-bentonite-water slurry sprayed onto pastures (26 kg MgO and 2.6 kg bentonite/ha) is effective in providing protection in high-rainfall periods. Provision in Drinking Water The problem with water medication is that the water intake of the group to be treated is not known and may be minimal on rapidly growing pastures. However, water medication may provide a delivery system for magnesium on management systems such as extensive range pastures where other methods may have limited success. Water sources other than the medicated supply need to be fenced off or otherwise restricted. The addition of magnesium sulfate (500 g/100 L) or magnesium chloride

1705

hexahydrate (420 g/100 L) to the water supply during the risk period for hypomagnesemia has proved effective. Management of Pasture Fields The economics of dairy farming make it necessary to produce maximum pasture growth, and the development of tetany-prone pastures is unavoidable in many circumstances. In some areas it may be possible to reduce the danger of such pastures by encouraging the development of legumes. In other areas the period of legume growth does not coincide with the period of maximum risk for grass tetany. Restricting the amount of potash added to pastures, especially in the period immediately preceding the risk period for tetany, or using potash fertilizers in the autumn or late spring after the period of risk can reduce risk of the disease. The grazing of low-risk animals on high-risk pastures is another strategy. Ensuring that ample salt is available during the danger period to counteract the high intake of potassium can also reduce risk of the disease. Plant geneticists are developing cultivars of cool-season grasses with high magnesium content that could be used for grazing during the tetany season. Lactating sheep grazing a high-magnesium cultivar of perennial rye grass (Lolium perenne cv Radmore) in the spring have shown higher serum magnesium concentrations than sheep grazing control cultivar, and cultivars of tall fescue (Festuca arundinacea) with high magnesium and calcium concentrations and low tetany potential are also available. Provision of Shelter In areas where winter pasturing is practiced, the observation that serum magnesium levels fall during the winter and in association with inclement weather suggests that cattle and sheep should be provided with shelter at such times. If complete housing is impractical, it may be advisable to erect open-access shelters in those fields that have no tree cover or protection from prevailing winds. Fields in which lactating cows are kept should receive special attention in this regard. Unfortunately, the disease is most common on highly improved farms, where most natural shelter has been removed, and it is desired to keep the cows on the highly improved pasture to maintain milk production or fatten calves rapidly. Time of Calving In areas where the incidence of the disease is high, it may be advisable to avoid having the cows calve during the cold winter months when seasonal hypomagnesemia is most likely to develop. Unfortunately, it is often important to have cows calve in late winter to take advantage of the flush of spring growth when the cows are at the peak of their lactation.

1706

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Feeding on Hay and Unimproved Pasture Because of the probable importance of lush, improved, grass pasture in producing the disease, the provision of some grain, hay, or rough grazing may reduce its incidence. It is most important that the periods of fasting, such as occur when cattle or sheep are yarded or moved or during bad weather, should be avoided, especially in lactating animals and when seasonal hypomagnesemia is likely to be present. TREATMENT AND CONTROL Treatment IV administration of a solution containing 25% calcium borogluconate and 5% magnesium hypophosphite (500 mL for cattle, 50 mL for sheep and goats) (R-1) SC administration of a 50% solution of magnesium sulfate (200 mL for cattle) (R-1) IV administration of a 15% solution of magnesium gluconate (200 to 400 mL for cattle) or a 20% solution of magnesium sulfate (200 to 300 mL for cattle) (R-2) Control Magnesium oxide—daily administration by drenching or in the feed of 60 g (cattle) or 7 g (sheep and goats) (R-1) Increase dietary magnesium intake at times of increased risk for hypomagnesemia (R-2) Monitor urine magnesium-to-creatinine ratio of animals at increased for hypomagnesemia (R-2)

FURTHER READING Brozos C, Mavrogianni VS, Fthenakis GC. Treatment and control of periparturient metabolic diseases: pregnancy toxemia, hypocalcemia, hypomagnesemia. Vet Clin North Am Food Anim Pract. 2011;27:105-113. Foster A, Livesy C, Edwards G. Magnesium disorders in ruminants. In Pract. 2007;29:534-539. Goff JP. Calcium and magnesium disorders. Vet Clin North Am Food Anim Pract. 2014;30:359-381. Martin-Tereso J, Martens H. Calcium and magnesium physiology and nutrition in relation to the prevention of milk fever and tetany (dietary management of macrominerals in preventing disease). Vet Clin North Am Food Anim Pract. 2014;30:643-670. Schonewille JT. Magnesium in dairy cow nutrition: an overview. Plant Soil. 2013;368:167-178.

REFERENCES

1. Edwards G, Foster A, Livesey C. In Pract. 2009;31: 22-25. 2. Schweigel M, et al. J Anim Physiol Anim Nutr. 2009;93:105-112.

HYPOMAGNESEMIC TETANY OF CALVES SYNOPSIS Etiology Hypomagnesemia, resulting from inadequate magnesium in the diet.

Epidemiology Most commonly calves 2 to 4 months of age, on whole milk or milk-replacer diets and poor or no roughage. Diarrhea and chewing of bedding or other coarse fiber may exacerbate the deficiency. Clinical findings Apprehension, agitation, hypersensitivity to all external stimuli, fine muscle tremors progressing to spasticity and violent convulsions. Rapid course and high case-fatality rate. Clinical pathology Serum magnesium concentrations below 0.8 mg/dL, bone calcium : magnesium ratio above 90 : 1. Necropsy findings Calcification of the spleen, diaphragm, and endothelium of the aorta and endocardium. Enzootic muscular dystrophy is often concurrent. Diagnostic confirmation Blood magnesium and response to treatment. Bone calcium : magnesium ratios. Treatment and control Magnesium injection and dietary supplementation with magnesium compounds.

ETIOLOGY The disease results when the dietary intake of magnesium is inadequate for the requirements of the calf. Affected animals may have concurrent hypocalcemia. Magnesium Homeostasis in the Calf Milk has low concentrations of magnesium. A milk diet provides adequate magnesium for the requirements of a growing calf up to a body weight of approximately 50 kg, but if milk is the sole diet, the intake of magnesium will be inadequate for requirements once this body weight is reached. The deficit will perpetuate if the other feeds that are fed are also low in magnesium. In the young calf, magnesium is absorbed in the intestine; however, the efficiency of magnesium absorption decreases from 87% to approximately 30% at 3 months of age, when maximum susceptibility to the disease occurs. The efficiency of absorption is also decreased by a reduction in intestinal transit time caused by diarrhea. In contrast to adult cattle, young calves can mobilize body stores of magnesium, which are principally located in the skeleton. Approximately 40% of the magnesium stored in the skeleton can be mobilized, which will protect against a short-term deficit. Hypomagnesemic tetany in calves is often complicated in field cases by the coexistence of other diseases, especially enzootic muscular dystrophy.

EPIDEMIOLOGY Occurrence The disease is not common. Cases may occur sporadically, or a number of deaths may occur on one farm within a short period of time.

Risk Factors The disease can occur under a number of different circumstances. Most commonly, hypomagnesemic tetany occurs in calves 2 to 4 months of age or older that are fed solely on a diet of whole milk, and calves receiving the greatest quantity of milk and growing most rapidly are more likely to be affected because of their greater need for magnesium for incorporation into developing soft tissues. It is most likely to occur in calves being fattened for veal. Those cases that occur in calves on milk replacer appear to be related to chronic scours and the low magnesium content of the replacer. This problem is less common than it once was because most modern commercial milk replacers have added adequate magnesium. A significant loss of magnesium in the feces also occurs in calves allowed to chew fibrous material, such as bedding; the chewing stimulates profuse salivation and creates greater loss of endogenous magnesium. Peat and wood shavings are bedding materials known to have this effect. Cases have also been reported in calves fed milk-replacer diets or milk, concentrates, and hay, and in calves running at pasture with their dams. Deaths resulting from hypomagnesemic tetany have also occurred in 3- to 4-month-old calves whose hay and silage rations were low in magnesium content. Hypomagnesemia also occurs in young cattle, about 6 months of age, that are being fattened intensively indoors for the baby beef market. The phosphorus content of their diet is high, and a lack of vitamin D is probable. The situation is exacerbated by a shortage of roughage. The hypomagnesemia is accompanied by hypocalcemia. Experimental Reproduction A condition closely resembling the field syndrome has been produced experimentally by feeding an artificial diet with a very low content of magnesium and a high calcium content, and biochemical hypomagnesemia is readily produced in calves with a diet based on skim milk and barley straw. Hypomagnesemia has also been produced experimentally in very young foals by feeding a diet with a very low magnesium content. The clinical signs are similar to those in calves, and the calcification found in the walls of vessels of calves also occurs in foals.

PATHOGENESIS

On affected farms, calves are born with normal serum magnesium concentrations of 2 to 2.5 mg/dL (0.82 to 1.03 mmol/L), but the concentrations fall gradually in the succeeding 2 to 3 months, often to below 0.8 mg/ dL (0.33 mmol/L). Tetany does not occur until the serum magnesium falls below this concentration and is most severe at concentrations below 0.6 mg/dL (0.25 mmol/L), although some calves in a group may have

Metabolic Diseases of Ruminants

VetBooks.ir

concentrations even lower than this and show few clinical signs. Magnesium deficiency inhibits the release and action of parathyroid hormone, and this is thought to be the genesis of the concurrent hypocalcemia. It is probable that depression of the serum calcium level precipitates tetany in animals rendered tetany prone by low serum magnesium levels. Tetanic convulsions can occur in hypocalcemic calves in the absence of hypomagnesemia. Hypomagnesemic tetany is not related in any way to enzootic muscular dystrophy, although the diseases may occur concurrently.

CLINICAL FINDINGS

The first sign in the experimental disease is constant movement of the ears. The temperature is normal and the pulse rate accelerated. Hyperesthesia to touch and grossly exaggerated tendon reflexes with clonus are present. Shaking of the head, opisthotonos, ataxia without circling, and a droopy, backward carriage of the ears are constant. There is difficulty in drinking because of the animal’s inability to get to the bucket. Initially, the calves are apprehensive, show agitation and retraction of the eyelids when approached, and are hypersensitive to all external stimuli, but they show no tetany. Later, fine muscle tremors appear, followed by kicking at the belly, frothing at the mouth, and spasticity of the limbs. Convulsions follow, beginning with stamping of the feet, head retraction, chomping of the jaws, and falling. During the convulsions the following signs are present: • Jaws are clenched. • Respiratory movements cease. • There are tonic and clonic movements of the limbs. • There is involuntary passage of urine and feces. • There are cycles of protrusion and retraction of the eyeballs. The pulse rate rises to 200 to 250/min, and the convulsions disappear terminally. The pulse becomes impalpable, and cyanosis appears before death. In field cases the signs are almost identical, but they are rarely observed until the terminal tetanic stage. Older calves usually die within 20 to 30 minutes of the onset of convulsions, but young calves may recover temporarily only to succumb to subsequent attacks. Cases that occur in young calves with scours, usually at about 2 to 4 weeks of age, show ataxia, hyperesthesia, opisthotonos, and convulsions as the presenting signs. The convulsions are usually continuous, and the calves die within 1 hour.

CLINICAL PATHOLOGY

Serum magnesium concentrations below 0.8 mg/dL (0.33 mmol/L) indicate severe hypomagnesemia, and clinical signs occur

with levels of 0.3 to 0.7 mg/dL (0.12 to 0.29 mmol/L). Normal values are 2.2 to −2.7 mg/dL (0.9 to 1.11 mmol/L). Erythrocyte magnesium concentrations are also low, indicating a chronic deficiency. Serum calcium concentrations tend to fall when serum magnesium levels become very low and are below normal in most clinical cases. The estimation of the magnesium in bone (particularly ribs and vertebrae) is a reliable confirmatory test at necropsy. Values below a ratio of 70 : 1 for calcium : magnesium may be regarded as normal, and those above 90 : 1 are indicative of severe magnesium depletion. In the normal calf the ratio is about 55 : 1.1 Absolute bone calcium values are not decreased and are often slightly elevated. An incidental change is the marked increase in serum creatinine kinase activity in calves after an acute attack of hypomagnesemic tetany.

NECROPSY FINDINGS

There is a marked difference between the necropsy lesions of some natural cases and those in the experimental disease. In field cases, there is often calcification of the spleen and diaphragm, and calcified plaques are present in the aorta and endocardium, together with hyaline degeneration and musculature. In other cases necropsy lesions similar to those in enzootic muscular dystrophy occur. In experimentally produced cases these lesions are not evident, but there is extensive congestion in all organs, and hemorrhages are present in unsupported organs, including the following: • Gallbladder • Ventricular epicardium • Pericardial fat • Aorta • Mesentery wall • Intestinal wall The lesions are obviously terminal and are associated with a terminal venous necrosis. Some field cases present a picture identical to this.

1707

Follow-up supplementation of the diet with magnesium oxide or carbonate as described later is advisable. Chloral narcosis or tranquilization with an ataractic drug may be essential to avoid death as a result of respiratory paralysis during convulsions.

CONTROL

The provision of hay that is high in magnesium, such as alfalfa, helps to prevent the disease, as will well-formulated concentrates. Supplementary Feeding of Magnesium If begun during the first 10 days of life, supplementary magnesium feeding will prevent excessive drops in serum magnesium, but if begun after the calf is 7 weeks old, it may not prevent further depression of the levels. Supplementation should continue until at least 10 weeks of age. Daily feeding of the magnesium compound and fairly accurate dosing are necessary to avoid scouring or inefficient protection. For calves of average growth rate, appropriate dose rates are 1 g/d for calves to 5 weeks of age, 2 g/d for calves 5 to 10 weeks of age, and 3 g/d for calves 10 to 15 weeks of age of magnesium oxide or twice this dose of carbonate. Supplementation of the diet with magnesium restores serum calcium levels to normal and corrects the hypomagnesemia. Magnesium Alloy Bullets Two bullets of the sheep size (together releasing approximately 1 g/d of magnesium) per calf have shown high efficiency in preventing the clinical disease and also the hypomagnesemia that precedes it. Calves kept indoors and fed largely on milk should get adequate mineral supplement and vitamin D (70,000 IU vitamin D3/d). Magnesium utilization will not be affected, but calcium absorption, which is often sufficiently reduced to cause a concurrent hypocalcemia, will be improved. REFERENCES

DIFFERENTIAL DIAGNOSIS • Acute lead poisoning • Enterotoxemia caused by Clostridium perfringens type D • Polioencephalomalacia • Tetanus • Vitamin A deficiency • Meningitis

TREATMENT

Response to magnesium injections (100 mL of a 10% solution of magnesium sulfate) is only transitory because of the severe depletion of bone reserves of magnesium. This dose provides only a single day’s requirements. A magnesium sulfate enema in warm water (containing 15 g of magnesium sulfate) was associated with a rapid response in hypomagnesemic 3-month-old calves.2

1. Foster A, et al. In Pract. 2007;29:534. 2. Soni AK, Shukla PC. Environ Ecol. 2012;30:1601.

TRANSPORT RECUMBENCY OF RUMINANTS Transport recumbency (tetany) occurs after prolonged transport, usually in cows and ewes in late pregnancy. It is also recorded in lambs transported to feedlots and in cows and sheep delivered to abattoirs. It is characterized by recumbency, alimentary tract stasis, and coma, and it is highly fatal. It occurs in most countries. Large losses can be encountered when cows and ewes in late pregnancy are moved long distances by rail, by truck, or on foot. Although cows of any age in late pregnancy are most commonly affected, the disease has also been recorded in cows

1708

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

recently calved, bullocks, steers, dry cows, and lambs. Risk factors include the following: • Heavy feeding before shipment • Deprivation of feed and water for more than 24 hours during transit • Unrestricted access to water • Exercise immediately after unloading There is an increased incidence of the disease during hot weather. The cause is unknown, although physical stress is an obvious factor. Lambs show the following characteristics: • Restlessness • Staggering • Partial paralysis of hindlegs • Early assumption of lateral recumbency Death may occur quickly, or after 2 to 3 days of recumbency. There is mild hypocalcemia (7 to 7.5 mg/dL; 1.75 to 1.87 mmol/L). The recovery rate even with treatment is only fair. Clinical signs may occur while the cattle are still on the transportation vehicle or up to 48 hours after unloading. In the early stages, animals may exhibit excitement and restlessness, trismus, and grinding of the teeth. A staggering gait with paddling of the hindlegs and recumbency occur, accompanied by stasis of the alimentary tract and complete anorexia. Animals that do not recover gradually become comatose and die in 3 to 4 days. There may be moderate hypocalcemia and hypophosphatemia in cattle. In sheep of various ages, some are hypocalcemic and hypomagnesemic, some are hypoglycemic, and some have no detectable biochemical abnormality. There are no lesions at necropsy other than those related to prolonged recumbency. Ischemic muscle necrosis is the most obvious of these lesions. The relationship of the disease to transport or forced exercise is diagnostic. Some cases respond to treatment with combined calcium, magnesium, and glucose injections. Repeated parenteral injections of large volumes of electrolyte solutions are recommended. In lambs, the SC injection of a solution of calcium and magnesium salts is recommended, but the response is usually only 50%, due probably because of an intercurrent myonecrosis. If prolonged transport of cows or ewes in advanced pregnancy is unavoidable, they should be provided with adequate food, water, and rest periods during the trip. The incidence of this condition after transportation appears to have been markedly reduced with increased monitoring and awareness of transportation-related morbidity and mortality.

KETOSIS AND SUBCLINICAL KETOSIS (HYPERKETONEMIA) IN CATTLE SYNOPSIS Etiology A multifactorial disorder of energy metabolism. Negative energy balance

results in hypoglycemia, ketonemia (the accumulation in blood of acetoacetate, β-hydroxybutyrate [BHB] and their decarboxylation products acetone and isopropanol), and ketonuria. Epidemiology Primary ketosis and subclinical ketosis occurs predominantly in wellconditioned cows with high lactation potential, principally in the first month of lactation, with a higher prevalence in cows with a higher lactation number. Loss of body condition in the dry period and immediately postpartum. Secondary ketosis occurs where other disease reduces feed intake. Clinical findings Cattle show wasting with decrease in appetite, body condition, and milk production. Some have short periods of bizarre neurologic and behavioral abnormality (nervous ketosis). Response to treatment is good. Subclinical ketosis (hyperketonemia) is detected by tests for ketones, usually BHB in blood, plasma, or serum, and acetoacetate in urine. Clinical pathology Hypoglycemia, ketonemia, ketonuria, or elevated ketones in milk. Necropsy findings None specific. Varying degrees of hepatic lipidosis. Diagnostic confirmation Ketonemia, ketonuria, or, less commonly, elevated ketone concentration in milk. Treatment Intravenous glucose, parenteral corticosteroid, and oral glucose precursors such as propylene glycol. The disease responds readily to treatment in cattle with mild hepatic lipidosis and is self-limiting. Control Correction of energy imbalance. Herd biochemical monitoring coupled with condition scoring. Daily monensin administration to late-gestation and early-lactation dairy cows.

ETIOLOGY Glucose Metabolism in Cattle The maintenance of adequate concentrations of glucose in the plasma is critical to the regulation of energy metabolism. The ruminant absorbs very little dietary carbohydrate as hexose sugar because dietary carbohydrates are fermented in the rumen to shortchain fatty acids, principally acetate (70%), propionate (20%), and butyrate (10%). Consequently, glucose needs in cattle must largely be met by gluconeogenesis. Propionate and amino acids are the major precursors for gluconeogenesis, with glycerol and lactate being of lesser importance. Propionate is produced in the rumen from starch, fiber, and proteins. It enters the portal circulation and is efficiently removed by the liver, which is the primary glucoseproducing organ. Propionate is the most important glucose precursor; an increased availability of propionate can spare the hepa­ tic utilization of other glucose precursors, and production of propionate is favored by a high grain inclusion in the diet. The

gluconeogenic effect of propionate should be contrasted to acetate, which is transported to peripheral tissues and to the mammary gland and metabolized to long-chain fatty acids for storage as lipids or secretion as milk fat. Amino acids. The majority of amino acids are glucogenic and are also important precursors for gluconeogenesis. Dietary protein is the most important quantitative source, but the labile pool of body protein (particularly skeletal muscle) is also an important source; together they contribute to energy synthesis , milk lactose synthesis, and milk protein synthesis. Energy Balance In high-producing dairy cows there is always a negative energy balance in the first few weeks of lactation. The highest dry matter intake does not occur until 8 to 10 weeks after calving, but peak milk production is at 4 to 6 weeks, and energy intake may not keep up with demand. In response to a negative energy balance and low serum concentrations of glucose (and consequently low serum concentrations of insulin), cows will mobilize adipose tissue, with consequent increases in serum concentrations of nonesterified fatty acids (NEFA) and subsequent increases in serum concentrations of β-hydroxybutyrate (BHB), acetoacetate, and acetone. The hepatic mitochondrial metabolism of fatty acids promotes both gluconeogenesis and ketogenesis. Cows partition nutrients during pregnancy and lactation and are in a lipolytic stage in early lactation and at risk for ketosis during this period. Hepatic Insufficiency in Ketosis Hepatic insufficiency has been shown to occur in bovine ketosis, but it does not occur in all cases. Ketosis is defined as an increased plasma or serum concentration of ketoacids and is divided into three types. In type I, or “spontaneous” ketosis, the gluconeogenic pathways are maximally stimulated, and ketosis occurs when the demand for glucose outstrips the capacity of the liver for gluconeogenesis because of an insufficient supply of glucose precursors. Rapid entry of NEFAs into hepatic mitochondria occurs and results in high rates of ketogenesis and high plasma/ serum ketone concentration. There is little conversion of NEFAs to triglycerides, resulting in little fat accumulation in the liver. In type II ketosis, manifest with fatty liver, gluconeogenic pathways are not maximally stimulated, and consequently mitochondrial uptake of NEFAs is not as active, and NEFAs become esterified in the cytosol, forming triglyceride. The capacity of cattle to transport triglyceride from the liver is low, resulting in accumulation and fatty liver. The occurrence of a fatty liver can further suppress hepatic gluconeogenic capacity. Hepatic insufficiency may occur more commonly in those cows predisposed to ketosis by overfeeding in the dry period. In type III ketosis, cattle

Metabolic Diseases of Ruminants

VetBooks.ir

are fed a diet (typically a high-maize ration) that results in a higher ruminal production of butyrate, which is directly metabolized by ruminal epithelial cells to butyrate. Ketone Formation Ketones arise from two major sources: butyrate in the rumen and mobilization of fat. A large proportion of butyrate produced by rumen fermentation of the diet is converted to BHB in the rumen epithelium and is absorbed as such. Free fatty acids produced from the mobilization of fat are transported to the liver and oxidized to produce acetylCoA and NADH. Acetyl-CoA may be oxidized via the tricarboxylic acid (TCA) cycle or metabolized to acetoacetyl-CoA. Complete oxidation of acetyl-CoA via the TCA cycle depends on an adequate supply of oxaloacetate from the precursor propionate. If propionate, and consequently oxaloacetate, is deficient, oxidation of acetyl-CoA via the TCA cycle is limited, and acetyl-CoA is metabolized to acetoacetyl CoA and subsequently to acetoacetate and BHB. The ketones BHB and acetoacetate can be utilized as energy sources. They are normally present in the plasma/serum of cattle, and their concentration is a result of the balance between production in the liver and utilization by the peripheral tissues. Acetoacetate can spontaneously convert to acetone, which is volatile and therefore exhaled in the breath; diffusion of acetone across the rumen epithelium into the rumen means that some acetone is eructated. Ruminal flora (most likely bacteria) can metabolize acetone to isopropanol, which can then be absorbed to increase plasma concentrations of isopropanol, a 3-carbon alcohol.1 Role of Insulin and Glucagon The regulation of energy metabolism in ruminants is primarily governed by insulin and glucagon. Insulin acts as a glucoregulatory hormone stimulating glucose use by tissues and decreasing hepatic gluconeogenesis. Plasma insulin concentrations decrease with decreasing plasma concentrations of glucose and propionate. Insulin also acts as a liporegulatory hormone stimulating lipogenesis and inhibiting lipolysis. Glucagon is the primary counterregulatory hormone to insulin. The counteracting effects of insulin and glucagon therefore play a central role in the homeostatic control of glucose. A low insulin : glucagon ratio stimulates lipolysis in adipose tissue and ketogenesis in the liver. Cows in early lactation have low insulin : glucagon ratios because of low plasma glucose concentrations and are in a catabolic state. Regulation is also indirectly governed by somatotropin, which is the most important determinant of milk yield in cattle and is also lipolytic. Factors that decrease the energy supply, increase the demand for glucose, or increase the utilization of peripheral fat

reserves as an energy source are likely to increase ketone production and ketonemia. There is, however, considerable cow-to-cow variation in the risk for developing clinical ketosis.

ETIOLOGY OF BOVINE KETOSIS

It is not unreasonable to view clinical ketosis as one end of a spectrum of a metabolic state that is common in heavily producing cows in the postcalving period. This is because high-yielding cows in early lactation are in negative energy balance and are subclinically ketotic as a result. Cattle are particularly vulnerable to ketosis because, although very little carbohydrate is absorbed as such, a direct supply of glucose is essential for tissue metabolism, particularly the formation of lactose associated with milk production. The utilization of volatile fatty acids for energy purposes is also dependent on a supply of available glucose. This vulnerability is further exacerbated in the lactating dairy cow by the tremendous rate of turnover of glucose. In the period between calving and peak lactation, the demand for glucose is increased and cannot be completely restrained. Cows will reduce milk production in response to a reduction of energy intake, but this does not follow automatically nor proportionately in early lactation because hormonal stimuli for milk production overcome the effects of reduced food intake. Under these cir­ cumstances, lowered plasma glucose con­ centrations result in lowered plasma insulin concentrations. Long-chain fatty acids are released from fat stores under the influence of both a low plasma insulin : glucagon ratio and the influence of high somatotropin concentration, and this leads to increased ketogenesis.

1709

ketosis, in addition to the importance of predisposing factors. Reviews of these studies are cited at the end of this disease section. In general, it can be stated that clinical ketosis occurs in cattle when they are subjected to demands on their resources of glucose and glycogen that cannot be met by their digestive and metabolic activity. A common classification of the disease based on its natural presentation in intensively and extensively managed dairy herds, and one that accounts for the early lactational demand for glucose, a limited supply of propionate precursors, and preformed ketones or mobilized lipids in the pathogenesis, has been developed. Such a classification scheme includes the following mechanisms for ketosis, which will be discussed in turn: • Primary ketosis (production ketosis) • Secondary ketosis • Alimentary ketosis • Starvation ketosis • Ketosis resulting from a specific nutritional deficiency Primary Ketosis (Production Ketosis) This is the ketosis of most herds, the socalled estate acetonemia. Primary ketosis occurs in cows in good to excessive body condition that have high lactation potential and are being fed good-quality rations but that are in a negative energy balance. There is a tendency for the disease to recur in individual animals, which is probably a reflection of variation between cows in digestive capacity or metabolic efficiency. A proportion of primary ketosis cases appear as clinical ketosis, but a much greater proportion occurs as cases of subclinical ketosis in which there are increased concentrations of circulating ketone bodies but no overt clinical signs. Affected cattle recover with correct feeding and ancillary treatment.

Individual Cow Variation The rate of occurrence of negative energy status, and therefore the frequency of clinical ketosis cases, has increased markedly over the last 4 decades because of the increase in the lactation potential of the modern dairy cow. Because of the mammary gland’s metabolic precedence in the partitioning of nutrients, especially glucose, milk production continues at a high rate, causing an energy drain. In many individual cows, the need for energy is beyond their capacity for dry matter intake, but there is between-cow variation in risk under similar nutritional stress. Clinical ketosis is easily produced in early-lactation dairy cows by reducing the daily feed intake.2 Subclinical ketosis (hyperketonemia) in early-lactation dairy cows is associated with decreased dry matter intake and feeding time during the week before calving.6

Secondary Ketosis Secondary ketosis occurs where the presence of other disease results in a decreased food intake. The cause of the reduction in food intake is commonly the result of abomasal displacement, traumatic reticulitis, metritis, mastitis, or other diseases common to the postparturient period. A high incidence of ketosis has also been observed in herds affected with fluorosis. An unusual occurrence reported was an outbreak of acetonemia in a dairy herd fed on a ration contaminated by a low level (9.5 ppm) of lincomycin, which caused ruminal microbial dysfunction. The proportion of cases of ketosis that are secondary and their diagnosis as such are both matters of great interest because a significant proportion of all cases of ketosis in lactating dairy cattle are secondary to other disease.

Types of Bovine Ketosis There are many theories on the cause and biochemical and hormonal pathogenesis of

Alimentary Ketosis Alimentary ketosis (also called type III in some classification systems) is a result of

1710

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

excessive amounts of butyrate in silage and possibly also a result of decreased food intake resulting from the poor palatability of high-butyrate silage. Silage made from succulent material may be more highly ketogenic than other types of ensilage because of its higher content of preformed butyric acid. Spoiled silage is also a cause, and toxic biogenic amines in silage, such as putrescine, may also contribute. This type of ketosis is commonly subclinical, but it may predispose to the development of production or primary ketosis. Starvation Ketosis Starvation ketosis occurs in cattle that are in poor body condition and that are fed poorquality feedstuffs. There is a deficiency of propionate and protein from the diet and a limited capacity of gluconeogenesis from body reserves. Affected cattle recover with correct feeding. Ketosis Resulting From Specific Nutritional Deficiency Specific dietary deficiencies of cobalt and possibly phosphorus may also lead to a high incidence of ketosis. This may be in part a result of a reduction in the intake of total digestible nutrients, but in cobalt deficiency, the essential defect is a failure to metabolize propionic acid in the TCA cycle. The problem is restricted to the cobalt-deficient areas of the world, although the occurrence of cobalt deficiency in high-producing dairy cows in nondeficient areas has been described. There is a marked nadir in food intake around calving, followed by a gradual increase. This increase is quite variable between cows, but in the great majority of cases does not keep pace with milk yield. The net result is that high-yielding dairy cows are almost certain to be in negative energy balance for the first 2 months of lactation.

EPIDEMIOLOGY Occurrence Ketosis is a very common disease of lactating dairy cattle and is prevalent in most countries where intensive farming is practiced. Ketosis occurs mainly in animals housed during the winter and spring months and is rare in cows that calve on pasture. In housed or free-stalled cattle, ketosis occurs year around. The occurrence of the disease is very much dependent on management and nutrition and varies between herds. As might be expected, lactational incidence rates vary between herds, and a review of 11 epidemiologic studies showed a lactation incidence rate for ketosis that varied from 0.2% to 10.0%. The incidence of subclinical ketosis (more correctly called hyperketonemia) is influenced by the cut-point of plasma BHB used for definition, but it is much higher than the incidence of clinical ketosis, especially in undernourished herds, and can

approach 40%. Incidence can be challenging and expensive to estimate because prevalence information is usually measured. In general, the incidence of subclinical ketosis is 1.8 times the prevalence.

areas, a higher risk is generally observed in cattle during the winter housing period. Higher prevalence has been observed in the late summer and early winter in Scandinavian countries.

Animal and Management Risk Factors There are conflicting reports on the significance of risk factors for ketosis and subclinical ketosis, which probably reflect that the disease can be a cause or effect of a number of interacting factors. The disease occurs in the immediate postparturient period, with 90% of cases occurring in the first 60 days of lactation. Regardless of the specific etiology, ketosis occurs most commonly during the first month of lactation, less commonly in the second month, and only occasionally in late pregnancy. In different studies, the median time to onset following calving has varied from 10 to 28 days, with some recent studies showing a peak prevalence of subclinical ketosis in the first 2 weeks postcalving. A prolonged previous intercalving interval increases risk.

Other Interactions.  There is a greater risk for the development of ketosis in cows that have an extended long dry period;3 those that develop milk fever, retained placenta, lameness, or hypomagnesemia; or those that have high milk production and high first milking colostrum volume.3 Cows with twins are also at risk for ketosis in the terminal stages of pregnancy. There is a bidirectional relation between risk for displaced abomasum and risk for ketosis, but in a field study of 1000 cows in 25 herds, cows that had a serum BHB concentration greater than 1.4 mmol/L in the first 2 weeks of lactation had odds of 4 : 1 that a displaced abomasum would be diagnosed 1 to 3 weeks later. In another study of 1010 cows, a serum BHB concentration of 1.5 mmol/L or greater in the first 2 weeks of lactation was found to be associated with a threefold increase in ketosis or displaced abomasum. Interestingly, cows with increased blood BHB concentration immediately before surgical correction of left-displaced abomasum have increased longevity within the herd, compared with cattle with BHB concentrations within the reference interval.7,8

Age.  Cows of any age may be affected, but the disease increases from a low prevalence at the first calving to a peak at the fourth calving, associated with the level of milk production. Lactational incidence rates of clinical ketosis of 1.5% and 9%, respectively, were found in a study of 2415 primiparous and 4360 multiparous cows. Clinical ketosis can also recur in the same lactation. Herd differences in prevalence are very evident in clinical practice and in the literature, with some herds having negligible occurrence. Although apparent differences in breed incidence are reported, evidence for a heritable predisposition within breeds is minimal. Feeding frequency has an effect, with the prevalence of ketosis being much lower in herds that feed a total mixed ration (TMR) ad libitum compared with herds that feed roughage and concentrate separately fed twice a day (component fed). Body-Condition Score.  There are conflicting reports on the relation between BCS at calving and ketosis, but it is very likely that studies that have found no relationship have not had many fat cows in the herds examined. Fat body condition postpartum was observed to be associated with a higher firsttest-day milk yield, milk-fat-to-protein ratio of greater than 1.5, increased body-condition loss, and a higher risk for ketosis. In another study, cows with a BCS greater than 3.25 at parturition and that lost 0.75 points in BCS in the first 2 months of lactation developed subclinical ketosis. Body-condition loss during the dry period also increases risk for ketosis in the following lactation. Season.  There is no clear association with season. In some but not all summer grazing

Economic Significance Clinical and subclinical ketosis are major causes of loss to the dairy farmer. In rare instances the disease is irreversible and the affected animal dies, but the main economic loss results from the loss of production while the disease is present, the possible failure to return to full production after recovery, and the increased occurrence of periparturient disease. Both clinical and subclinical ketosis are accompanied by decreased milk yields; lower milk protein and milk lactose; increased risk for delayed estrus and lower first-service conception rates; lower pregnancy rates; increased intercalving intervals; increased risk of cystic ovarian disease, metritis, and mastitis; and increased involuntary culling.4 The estimated economic loss from a single case of subclinical ketosis was US$117 in 2015, and the estimated average total cost per case of subclinical ketosis was $289 after considering the costs of displaced abomasum and metritis attributed to hyperketonemia.5

PATHOGENESIS Bovine Ketosis The principal metabolic disturbances observed, hypoglycemia and ketonemia, may both exert an effect on the clinical syndrome. However, in the experimental disease in cattle, it is not always clear what determines the development of the clinical signs in cases that convert from subclinical to clinical

Metabolic Diseases of Ruminants

VetBooks.ir

ketosis. In many cases, the severity of the clinical syndrome is proportional to the degree of hypoglycemia, and this, together with the rapid response to parenterally administered glucose in cattle, suggests hypoglycemia as the predominant factor. This hypothesis is supported by the development of prolonged hypoglycemia and a similar clinical syndrome to that of ketosis, after the experimental IV or SC injection of insulin (2 U/kg BW). However, in most field cases the severity of the clinical syndrome is also roughly proportional to the degree of ketonemia. This is an understandable relationship because ketone bodies are produced in larger quantities as the deficiency of glucose increases. However, the ketone bodies are thought to exert an additional influence on the clinical signs observed; for instance, acetoacetic acid is known to be toxic and probably contributes to the terminal coma in diabetes mellitus in humans. The nervous signs that occur in some cases of bovine ketosis are thought to be caused by the production of isopropanol, a breakdown product of acetone in the rumen,1 although the requirement of nervous tissue for glucose to maintain normal function may also be a factor in these cases. A reasonable explanation for the development of nervous ketosis is that a rapid increase in plasma acetone concentration in an animal that has an active rumen flora leads to a rapid increase in ruminal acetone concentration. The acetone is metabolized by rumen microflora to isopropanol, which is then absorbed into the bloodstream, potentially leading to neurologic abnormalities. This mechanism is consistent with observations that nervous signs of ketosis are more common in cattle with severe ketosis that is rapidly induced. Spontaneous ketosis in cattle is usually readily reversible by treatment; incomplete or temporary response is usually a result of the existence of a primary disease, with ketosis present only as a secondary development, although fatty degeneration of the liver in protracted cases may prolong the recovery period. Changes in ruminal flora after a long period of anorexia may also cause continued impairment of digestion. Immunosuppression has been demonstrated with energy deficiency and ketosis. The higher susceptibility of ketotic postpartum cows to local and systemic infections may be related to impairment of the respiratory burst of neutrophils that occurs with elevated plasma concentrations of BHB.

The wasting form is the most common of the two and is manifest with a gradual but moderate decrease in appetite and milk yield over 2 to 4 days. In component-fed herds, the pattern of appetite loss is often very specific in that the cow first refuses to eat grain, then ensilage, but may continue to eat hay. The appetite may also be depraved. Body weight is lost rapidly, usually at a greater rate than one would expect from the decrease in appetite. Farmers usually describe affected cows as having a “woody” appearance because of the apparent wasting and loss of cutaneous elasticity presumably resulting from disappearance of subcutaneous fat. The feces are firm and dry, but serious constipation does not occur. The cow is moderately depressed and is quieter than usual. The disinclination to move and to eat may suggest the presence of mild abdominal pain, but localized pain cannot be detected via abdominal palpation. The temperature and the pulse and respiratory rates are normal, and although the ruminal movements may be decreased in amplitude and number, they are within the normal range unless the course is of long duration, in which case they may virtually disappear. The characteristic sweet odor of ketones is detectable on the breath and often in the milk, but people vary in their ability to detect ketones on the breath (specifically the volatile ketone, acetone). Very few affected animals die, but without treatment the milk yield falls; although spontaneous recovery usually occurs over about a month, as equilibrium between the drain of lactation and food intake is established, the milk yield is never fully regained. The fall in milk yield in the wasting form may be as much as 25%, and there is an accompanying sharp drop in the solids-not-fat content of

the milk. In the wasting form, nervous signs may occur in a few cases, but they rarely comprise more than transient bouts of staggering and partial blindness. In the nervous form (nervous ketosis), signs are usually bizarre and begin quite suddenly. The syndrome is suggestive of delirium rather than of frenzy, and the characteristic signs include the following: • Walking in circles • Straddling or crossing of the legs • Head pushing or leaning into the stanchion • Apparent blindness • Aimless movements and wandering • Vigorous licking of the skin and inanimate objects (Fig. 17-6) • Depraved appetite • Chewing movements with salivation Hyperesthesia may be evident, with the animal bellowing on being pinched or stroked. Moderate tremor and tetany may be present, and there is usually an incoordinate gait. The nervous signs usually occur in short episodes that last for 1 or 2 hours and may recur at intervals of about 8 to 12 hours. Affected cows may injure themselves during the nervous episodes. Surgical correction of displaced abomasum in cows exhibiting some signs consistent with nervous ketosis should be delayed until their energy status has been evaluated and treatment instituted, if indicated. Subclinical Ketosis (Hyperketonemia) Subclinical ketosis is defined as an increase in blood/plasma/serum BHB above the normal reference range or ketonuria in a cow without detectable clinical signs of disease. Many cows that are in negative energy balance in early pregnancy will have ketonuria without showing clinical signs, but they

CLINICAL FINDINGS

Two major clinical forms of bovine ketosis are described—wasting and nervous—but these are the two extremes of a range of syndromes in which wasting and nervous signs are present in varying degrees of prominence.

1711

Fig. 17-6  Holstein–Friesian cow with nervous ketosis, manifest as excessive and sustained licking behavior.

1712

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

will have diminished productivity, including depression of milk yield and a reduction in fertility. Clinical diagnosis is not effective, and in one study, diagnosis by routine urine testing at 5 to 12 days postpartum was considerably more efficient (15.6% detected) than diagnosis by the herdsman (4.4% detected). In a British study of 219 herds the annual mean rate of reported clinical ketosis was 0.5 per 100 adult cows, but the rate of subclinical ketosis, as defined by increased plasma concentrations of BHB and nonesterified fatty acids, was substantially higher. There is debate about whether subclinical ketosis is the correct term, with some support for replacing the term with hyperketonemia. Potential milk production in cows with subclinical ketosis is reduced by 1% to 9%. Surveys of large populations show a declining prevalence of ketosis-positive cows after a peak in the period immediately after calving and a positive relationship between hyperketonemia and high milk yield. Infertility may appear as an ovarian abnormality, delayed onset of estrus, or endometritis resulting in an increase in the calving-toconception interval and reduced conception rate at first insemination.4

CLINICAL PATHOLOGY

Hypoglycemia, ketonemia, and ketonuria are characteristic of the disease. Glucose Plasma glucose concentrations are reduced from the normal of approximately 50 to 65 mg/dL to 20 to 40 mg/dL. Ketosis secondary to other diseases is usually accompanied by plasma glucose concentrations above 50 mg/dL, and many cattle have much higher concentrations. Conversion factors are shown in Table 17-7. Ketones Most commonly, plasma or serum βhydroxybutyrate (BHB) measured in SI units (mmol/L) is used for analysis of ketonemia. BHB is the quantitatively highest circulating ketone body in cattle. Plasma concentrations of BHB significantly correlate with plasma concentrations of acetoacetate, but acetoacetate is unstable in blood samples, whereas BHB is stable, particularly when samples are refrigerated or frozen. Normal cows have plasma BHB concentrations less

than 1.0 mmol/L; cows with subclinical ketosis have blood or plasma/serum concentrations greater than 1.0, 1.2, or 1.4 mmol/L (the cut-point varies depending on the study, analytical method, and whether blood or plasma is analyzed).9,10 Different cut-points have been proposed for serum BHB concentration in the first week postpartum (1.0 mmol/L) and the second week postpartum (1.4 mmol/L);11 this may be attributable to blood BHB concentrations being highest at 8 days in milk.12 In general, because the cut-point for the diagnosis of subclinical ketosis should be based on a detectable effect on decreasing milk production or an increased risk of adverse health events,10 a consensus is developing around the use of serum/plasma BHB concentration greater than 1.0 mmol/L as the cut-point for subclinical ketosis based on the association with impaired reproductive performance11 and increased risk of developing a displaced abomasum, puerperal metritis, or clinical ketosis.13 Cows with clinical ketosis usually have serum/plasma BHB concentrations in excess of 2.5 mmol/L, with values rarely reaching 10.0 mmol/L. Plasma BHB shows some diurnal variation in cows fed twice daily, with peak concentrations occurring approximately 4 hours after feeding and higher concentrations in the morning than in the afternoon. This diurnal variation is not as prominent in cows fed a total mixed ration ad libitum. Measurement of blood or plasma/serum BHB concentration has recently become a cost-effective and convenient method for routine analysis and cow-side monitoring, with the introduction of low-cost point-ofcare devices for measurement (US$2/test). The concentration of acetoacetate or BHB in urine and milk is also used for diagnostic purposes.14 Concentrations of BHB and acetoacetate in urine and milk are less than those in plasma/serum, but the correlation coefficients for plasma/serum and milk BHB and plasma/serum and milk acetoacetate are 0.66 and 0.62, respectively. For cow-side use, urine acetoacetate concentration using the nitroprusside test and blood BHB concentration using a point-of-care device are currently the preferred tests for detecting subclinical or clinical ketosis in cattle.

Table 17-7  To convert from the SI unit to the conventional unit, divide by the conversion factor; to convert from the conventional unit to the SI unit, multiply by the conversion factor Substrate

Conventional unit

β-hydroxybutyrate Acetoacetate Acetone

mg/dL mg/dL mg/dL

Conversion factor

SI unit

0.0961 0.0980 0.1722

mmol/L mmol/L mmol/L

Milk and Urine Cow-Side Tests Cow-side tests have the advantage of being inexpensive and giving immediate results, and they can be used as frequently as necessary. A minor source of error is that the concentration of ketone bodies in these fluids will depend not only on the ketone concentration of the plasma, but also on the amount of urine excreted or on the milk yield. Milk concentration of ketones is less variable, easier to collect, and may give fewer false negatives in cows with subclinical ketosis. Milk and urine ketone concentrations have been traditionally detected by the reaction of acetoacetate with sodium nitroprusside and can be interpreted in a semiquantitative manner based on the intensity of the reaction. The nitroprusside reaction detects both acetoacetate and acetone, but it is much more sensitive to acetoacetate than acetone; the latter is only detected when acetone concentrations are greater than 600 mmol/L, which represents a supraphysiologic concentration.15 As a consequence, the nitroprusside test functions as a semiquantitative test of acetoacetate concentration and should be clinically regarded as a test of acetoacetate and not acetone. Several products are available commercially as strips or test powders and are commonly accom­ panied by a color chart that allows a clas­ sification of acetoacetate concentration in grades such as negative, trace (5 mg/dL; 0.5 mmol/L), small (15 mg/dL; 1.0 mmol/L), moderate (40 mg/dL; 2.0 mmol/L), or large (>80 mg/dL; 5 mmol/L), based on the intensity of the color of the reaction.15 Milk powder tests are not sufficiently sensitive for detection of subclinical ketosis (report too many false negatives), and urine tests are not sufficiently specific (report too many false positives). Milk Testing.  The sensitivity and specificity of the nitroprusside powder test with milk in various studies is reported as 28% to 90% and 96% to 100%, respectively. Currently, a milk strip test detecting the concentration of BHB in milk is available and is graded on the concentration of BHB. In different studies, milk BHB has a reported sensitivity and specificity of 58% to 96% and 69% to 99%, respectively. These variations are in part a result of the use of different plasma BHB reference values (1.2 and 1.4 mmol/L) for designation of subclinical ketosis and different statistical methods for analysis. Somatic cell counts in milk greater than 1 million cells/mL will cause an elevation in reading of both the BHBA strip test and the nitroprusside tests. Urine Testing.  A nitroprusside tablet has a reported sensitivity and specificity of 100% and 59%, respectively, compared with serum BHB concentrations above 1.4 mmol/L; a nitroprusside strip test has a reported

Metabolic Diseases of Ruminants

VetBooks.ir

sensitivity and specificity of 78% and 96%, respectively, with a urine cut-point corresponding to “small” on the color chart or 49% and 99%, respectively, with a urine cut-point corresponding to “moderate” on the color chart. BHB test strips when used with urine have a reported sensitivity and specificity of 73% and 96%, respectively, at a urine cut-point of 0.1 mmol/L BHB and 27% and 99%, respectively, at a urine cutpoint of 0.2 mmol/L BHB. Urinary ketone concentrations are more closely related to plasma ketone concentrations than are milk BHB and acetoacetate concentrations.16,17 Moreover, urine acetoacetate concentration appears superior to milk BHB concentration in diagnosing ketosis.17 Milk-Fat-to-Protein Ratio.  Milk-fat concentration tends to increase, and milk protein concentration tends to decrease, during postpartum negative energy balance. A fatto-protein ratio greater than 1.5 in first-day test milk is indicative of a lack of energy supply in the feed and of risk for ketosis and provides a similar test sensitivity (Se = 0.63) for detecting subclinical ketosis as does milk BHB concentration (Se = 0.58).17 Milk production in multiparous animals is also separately associated with postpartum negative energy balance.18 Clinical Chemistry and Hematology.  White and differential cell counts are variable and not of diagnostic value for ketosis. There are usually elevations of liver enzyme activity in plasma/serum, but liver function tests are within the normal range. Liver biopsy is the only accurate method to determine the degree of liver damage. Plasma concentrations of NEFAs and total bilirubin are elevated in ketosis, with mean NEFA concentrations increasing above 0.3 mmol/L from 3 days before parturition to approximately 0.7 mmol/L from 0 to 9 days in milk, after which time plasma NEFA concentration gradually decreases.12 The increase in bilirubin is attributed, in part, to hepatic dysfunction; however, bilirubin is not a sufficiently sensitive indicator to assess the extent of fat mobilization and liver function in cows with ketosis. Plasma cholesterol concentration is typically decreased for the stage of lactation; the decrease in cholesterol is a result of decreased hepatocyte secretion of very-low-density lipoproteins (VLDLs), which are cholesterol rich, or increased mammary uptake of cholesterol relative to cholesterol availability. After secretion, VLDLs are processed in plasma to intermediate-density lipoproteins by hydrolysis of triglycerides.19 Intermediate-density lipoproteins are then metabolized in plasma to cholesterol-rich low-density lipoproteins that carry cholesterol to peripheral tissues, including the mammary gland.19,20 A clinically significant proportion of lactating dairy cattle with ketosis have low plasma cortisol

concentrations;21 although the mechanism has not been determined, it is possible that decreased cholesterol availability negatively affects cortisol synthesis. Liver glycogen levels are low, and the glucose tolerance curve may be normal. Volatile fatty acid levels in the rumen are much higher in ketotic than in normal cows, and the ruminal concentrations of butyrate are markedly increased relative to acetate and propionate acids. There is a small but significant drop in serum calcium concentrations (down to about 9 mg/dL [2.25 mmol/L]), probably as a result of decreased dry matter intake in lactating dairy cattle relative to the level of milk production. Plasma and urine metabolic profiling shows promise as a means of differentiating cattle with clinical ketosis and subclinical ketosis from healthy cattle at the same stage of lactation. Twenty-five plasma metabolites22,23 and 11 urine proteins24 have been identified to differ between these three groups. Differences include changes in plasma amino acid concentrations that may reflect differences in feed intake relative to milk production or altered metabolic pathways and changes in urine polypeptide concentrations that may reflect decreased immune responsiveness.

NECROPSY FINDINGS

The disease is not usually fatal in cattle, but fatty degeneration of the liver and secondary changes in the anterior pituitary gland and adrenal cortex may be present. DIFFERENTIAL DIAGNOSIS Cattle The clinical picture is usually too indefinite, especially in cattle, to enable a diagnosis to be made solely on clinical grounds. General consideration of the history, with particular reference to the time of calving, and the feeding program, and biochemical examination to detect the presence of hypoglycemia, ketonemia, and ketonuria are necessary to establish a diagnosis. Wasting form:

• • • •

Abomasal displacement Traumatic reticulitis Primary indigestion Cystitis and pyelonephritis

Nervous form: • Rabies • Hypomagnesemia • Bovine spongiform encephalopathy

TREATMENT In cattle, a number of effective treatments are available for ketosis, but in some affected animals, the response is only transient; in rare cases, the disease may persist and cause death or necessitate slaughter of the animals. Most of these cases are secondary, and failure to respond satisfactorily to treatment is a result of the primary disease. Specific

1713

treatment for subclinical ketosis is usually not applied on an individual basis, but nutrition and management issues should be investigated whenever a large proportion of early-lactation cows are diagnosed with subclinical ketosis. The rational treatment in ketosis is to relieve the need for glucose formation from tissues and allow ketone-body utilization to continue normally. Theoretically, the simplest means of doing this is by the administration of glucose replacement therapy. The effect of the administration of glucose is complex, but it allows the reversal of ketogenesis and the establishment of normal patterns of energy metabolism. Ideally, treatment should be at an early stage of the disease to minimize loss, and with subclinical ketosis this requires biochemical testing. Replacement Therapy Glucose (Dextrose) The IV injection of 500 mL of a 50% solution of glucose results in transient hyperglycemia, increased insulin and decreased glucagon secretion, and reduced plasma concentration of NEFAs. Glucose administration effects a marked improvement in most cows, but relapses occur commonly unless repeated treatments are used. This is probably a result of the transience of the hyperglycemia (3 to 4 hours) or insufficient dosing—the dose required varies directly with the amount of lactose being lost in the milk. Contrary to widespread belief, very little of the administered glucose is lost to urinary excretion (14 days) to dairy cattle in the United States at 185 to 660 (mg/head)/ day monensin to lactating cows or 115 to 410 (mg/head)/day monensin to dry cows. To accomplish this, monensin is approved to be fed at 11 to 22 g/ton of total mixed ration on a 100% dry matter basis, at a daily per-cow cost of about 2 to 4 cents. Monensin is also approved for use in the United States as part of a component feeding system at 11 to 400 g/ton (as is basis); this includes application as a “top dress,” where a small amount of feed is added to a ration. In some countries, monensin can be administered orally as a controlled-release capsule to cattle 2 to 4 weeks before calving. The capsule contains 32 g of monensin and releases approximately 335 mg monensin a day for 95 days. This product is effective and practical for a variety of feeding systems, and approximately 18% of dairy herds in Canada are administering monensin by controlledrelease capsule. Corticosteroids Isoflupredone acetate (20 mg, IM, once) was not effective in preventing subclinical ketosis in early-lactation dairy cows, and it actually increased the likelihood of subclinical ketosis.34 Ancillary Agents A commercially available injectable product containing cyanocobalamin (vitamin B12, 1 to 4 mg daily IV) in a combined formulation with butaphosphan is effective in normalizing energy status when administered to dairy cattle 2 to 6 times before or around parturition.29,30 The administration of cyanocobalamin and butaphosphan may be most beneficial in cows at increased risk of developing ketosis, such as older cows, overconditioned cows, or those experiencing dystocia or metritis.29 Phosphorus may be limiting in early lactation, based on low liver phosphorus content in dairy cattle.35 It is not clear whether additional phosphorus mitigates the reduction in hepatic phosphorus content. Rumen protected choline (15 g/day) fed daily starting 25 days before calving and continuing to 80 days after calving decreased the incidence of clinical ketosis and improved the health of lactating dairy cows.36 Choline

1716

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

is a precursor for phosphatidylcholine, which is thought to be rate limiting in early lactation; phosphatidylcholine deficiency is associated with impaired lipid metabolism. Niacin is antilipolytic and induces increases in blood glucose and insulin, but there is conflicting evidence that niacin given in the feed has a beneficial effect on subclinical ketosis in cattle. It has been suggested that niacin should be supplemented from 2 weeks before parturition to 12 weeks postpartum. General Control Herd Monitoring.  There is currently no consensus as to the optimal monitoring program for ketosis and subclinical ketosis in lactating dairy cattle, and consequently a variety of monitoring programs have been proposed. Challenges with developing optimal monitoring programs are the herd size (through the influence on the eligible numbers of animals available to be tested), ease of testing, cost of the test, and test sensitivity and specificity. In addition, the goals of the monitoring program need to be defined; typically they are either to monitor the adequacy of the diet relative to the level of milk production (i.e., the magnitude of negative energy balance in early lactation) or to identify animals to receive a standard treatment protocol, such as daily oral propylene glycol drenching. The optimal time for testing appears to be cows 3 to 9 days in milk because cows that are hyperketonemic at this stage of lactation are at highest risk for subsequent negative production and health effects, with the incidence and prevalence of subclinical ketosis occurring on day 5 of lactation.37 A recent modeling approach utilizing 13,000 cows from 833 dairy farms in North America and Europe suggested that testing cows twice weekly from 3 to 9 days in milk was the most cost effective strategy when the subclinical ketosis incidence was between 15% and 50%; below an incidence of 15% it was not economical to test, and above 50% all cows should be treated without testing.38 In addition, whenever the subclinical ketosis incidence increased to above 15%, a variety of testing and treatment protocols are economically beneficial.38 The six most valuable and practical indices for monitoring negative energy balance are urine acetoacetate concentration, blood BHB concentration, blood glucose concentration, body-condition score, backfat thickness determined ultrasonographically, and milk-fat-to-protein ratio. The first five indices can be obtained cow side and at no cost or relatively low cost, although determining the blood BHB concentration costs approximately 5 to 10 times that of the first two tests and requires a blood sample. The milk-fat-to-protein ratio is readily obtained from individual monthly test data and is more highly correlated with energy balance than plasma BHB or glucose concentration.39

This should be coupled with body-condition scoring or back-fat thickness to monitor the efficacy of the nutritional program. Plasma NEFA concentration is an excellent monitoring test of negative energy balance, but it is currently too expensive for routine herd monitoring, and an easy-to-use cow-side test is not available. Urine testing using the nitroprusside test for acetoacetate is the simplest of the cow-side tests, and despite some reports that urine samples are difficult to obtain from all cattle, urine is easily obtained from more than 90% of cattle using the following standardized technique. First, stimulation of the perineum to obtain a urine sample must be the first part of the examination of the cow and ideally should be performed without the cow being aware that the veterinarian is present. Second, never hold the tail while stimulating the perineum because tail holding alerts the cow to the presence of the veterinarian, and it is not needed because cattle never urinate on their tails when posturing to urinate. Third, obtain urine samples in the normal environment of the animal; because cattle urinate on average five times per day, urine samples are easily obtained on recumbent cattle that are gently encouraged to stand. Blood BHB testing has become very popular because of the availability of lowcost point-of-care meters. Despite this, it must be recognized that obtaining a blood sample is more complicated than obtaining a urine sample, and that the cost, although low, is much higher than that for urine acetoacetate or blood glucose testing. Moreover, serum BHB concentration is correlated with energy balance in a similar manner to plasma glucose concentration.40 Automated monitoring by in-line measurements of ketone bodies in milk has been studied and may be of particular value in large dairies. BHB is proposed as the candidate because it is the more robust in milk, and where cows are fed a total mixed ration, it is not subject to significant diurnal variation. Milk BHB concentration can be measured in real-time with a fluorometric method that requires no pretreatment of the milk. Biochemical monitoring of herds for subclinical ketosis and adequacy of periparturient feeding can be conducted using blood glucose estimations on a sample of cows in their second week of lactation. Plasma glucose concentrations below 45 mg/dL (2.4 mmol/L) suggest subclinical ketosis. For individual cows, blood glucose estimations should be done at about 14 days after calving. This method of monitoring is inexpensive using widely available point-of-care devices.

adaptations around calving and strategies to reduce feeding-related diseases. Anim Feed Sci Technol. 2006;126:175-213. McArt JAA, Nydam DV, Oetzel GR, Overton TR, Ospina PA. Elevated non-esterified fatty acids and β-hydroxybutyrate and their association with transition dairy cow performance. Vet J. 2013;198:560-570. Opsina PA, McArt JA, Overton TR, Stokol T, Nydam DV. Using nonesterified fatty acids and β-hydroxybutyrate concentrations during the transition period for herd-level monitoring of increased risk of disease and decreased reproductive and milking performance. Vet Clin North Am Food Anim Pract. 2013;29:387-412. Zhang Z, Liu G, Wang H, Li X, Wang Z. Detection of subclinical ketosis in dairy cows. Pakistan Vet J. 2012;32:156-160.

REFERENCES

1. Sato H. Anim Sci J. 2009;80:381. 2. Loor JJ, et al. Physiol Genomics. 2007;32:105. 3. Vanholder T, et al. J Dairy Sci. 2015;98:880. 4. Shin EK, et al. Theriogenology. 2015;84:252. 5. McArt JAA, et al. J Dairy Sci. 2015;98:2043. 6. Goldhawk C, et al. J Dairy Sci. 2009;92:4971. 7. Croushore WS, et al. J Am Vet Med Assoc. 2013;243:1329. 8. Reynen JL, et al. J Dairy Sci. 2015;98:3806. 9. Kessel S, et al. J Anim Sci. 2008;86:2903. 10. Borchardt S, et al. J Am Vet Med Assoc. 2012;240:1003. 11. Walsh RB, et al. J Dairy Sci. 2007;90:2788. 12. McCarthy MM, et al. J Dairy Sci. 2015;98:6284. 13. Opsina PA, et al. J Dairy Sci. 2010;93:546. 14. Denis-Robichaud J, et al. Bovine Pract. 2011;45:97. 15. Smith SW, et al. Acad Emerg Med. 2008;15:751. 16. Larsen M, Kristensen NB. Acta Agric Scand Sect A. 2010;60:239. 17. Krogh MA, et al. J Dairy Sci. 2011;94:2360. 18. Kayano M, Kataoka T. J Vet Med Sci. 2015;in press. 19. Kessler EC, et al. J Dairy Sci. 2014;97:5481. 20. Gross JJ, et al. PLoS ONE. 2015;10(6):doi:10.1371. 21. Forslund KB, et al. Acta Vet Scand. 2010;52:31. 22. Sun LW, et al. J Dairy Sci. 2014;97:1552. 23. Li Y, et al. Vet Quart. 2014;54:152. 24. Xu C, et al. Vet Quart. 2015;35:133. 25. Grunberg W, et al. J Vet Intern Med. 2006;20:1471. 26. Grunberg W, et al. J Dairy Sci. 2011;94:727. 27. Kusenda M, et al. J Vet Intern Med. 2013;27:200. 28. Djokovic R, et al. Acta Vet Brno. 2007;76:533. 29. Rollin E, et al. J Dairy Sci. 2010;93:978. 30. Furll M, et al. J Dairy Sci. 2010;93:4155. 31. Duffield TF, et al. J Dairy Sci. 2008;91:1334. 32. Duffield TF, et al. J Dairy Sci. 2008;91:1347. 33. Duffield TF, et al. J Dairy Sci. 2008;91:2328. 34. Seifi H, et al. J Dairy Sci. 2007;90:4181. 35. Grunberg W, et al. J Dairy Sci. 2009;92:2106. 36. Lima FS, et al. Vet J. 2012;193:140. 37. McArt JAA, et al. J Dairy Sci. 2012;95:5056. 38. McArt JAA, et al. Prev Vet Med. 2014;117:170. 39. Reist M, et al. J Dairy Sci. 2002;85:3314.

FURTHER READING

FATTY LIVER IN CATTLE (FAT-MOBILIZATION SYNDROME, FAT-COW SYNDROME, HEPATIC LIPIDOSIS, PREGNANCY TOXEMIA IN CATTLE)

Gordon JL, LeBlanc SJ, Duffield TF. Ketosis treatment in lactating dairy cattle. Vet Clin North Am Food Anim Pract. 2013;29:433-445. Ingvartsen KL. Feeding- and management-related diseases in the transition cow. Physiological

Fatty liver (hepatic lipidosis) is an important metabolic disease of dairy cows in early lactation and is associated with decreased health status and reproductive performance.

Metabolic Diseases of Ruminants

VetBooks.ir

SYNOPSIS Etiology Mobilization of excessive body fat to liver during periods of negative energy balance at time of parturition or in early lactation of dairy cows and late pregnancy of beef cows. Epidemiology High-producing dairy cows overfed during dry period may develop fatty liver syndrome just before or after calving precipitated by any factor or disease that interferes with feed intake. Occurs in well-conditioned beef cattle in late pregnancy when energy intake is suddenly decreased. Moderate and subclinical degrees of fatty infiltration may adversely affect reproductive performance of dairy cows. Signs Inappetence to anorexia, ruminal atony, lethargic, inactivity, ketonuria, fat body condition, weakness and recumbency if worsens. Recover if continue to eat and appetite improves. Clinical pathology Increase in plasma/serum nonesterified fatty acid, acetoacetate, β-hydroxybutyrate, and total bilirubin concentrations; increase in plasma/serum hepatic enzyme activity (particularly aspartate aminotransferase and ornithine carbamoyl transferase activity); increased fat content in liver biopsy. Necropsy findings Fatty infiltration of liver, liver may appear yellow. Diagnostic confirmation Liver biopsy. Differential diagnosis list • Left-sided or right-sided displacement of abomasum • Milk fever • Abomasal impaction • Vagus indigestion • Peritonitis Treatment Fluid and electrolyte therapy including glucose IV (bolus infusion). Propylene glycol orally. Dexamethasone IM. Provision of palatable feed. Control Avoid overfeeding during late lactation and dry period. Avoid situations that reduce feed intake at time of parturition.

ETIOLOGY Fatty liver is caused by the mobilization of excessive quantities of fat from body deposits to the liver. It develops when the hepatic uptake of lipids exceeds the oxidation and secretion of lipids by the liver. Excess lipids are stored as triacylglycerol in the liver, and excessive lipid in hepatocytes is associated with decreased metabolic function of the liver. Fatty liver occurs because of a sudden demand of energy in the immediate postpartum period in well-conditioned lactating dairy cows. Fatty liver also occurs because of a sudden deprivation of feed in fat pregnant beef cattle, and is especially severe in those bearing twins. The disease is an exaggeration

of what is a common occurrence in highproducing dairy cows that are in a state of negative energy balance in early lactation. A substantial drop in voluntary dry matter intake is initiated in late pregnancy and continues into early lactation. This decrease has traditionally been interpreted as caused by physical constraints in the abdomen as a result of the enlarging gravid uterus, but this purported mechanism appears to have been overemphasized. The decline in dry matter intake coincides with changes in reproduction status, changes in fat mass, and metabolic changes in support of lactation, and the associated metabolic signals are likely to play an important role in intake regulation. These signals include nutrients, metabolites, reproductive hormones, stress hormones, leptin, insulin, gut peptides, cytokines, and neuropeptides. Body fat, especially subcutaneous fat, is mobilized and deposited primarily in liver but also in muscle and the kidneys. Whether or not the cow is truly fat at parturition may not be important in determining the degree of fat mobilization, but the degree of negative energy balance in early lactation is critical.

EPIDEMIOLOGY Occurrence and Incidence Fatty infiltration of the liver is common in high-producing dairy cattle from a few weeks before and after parturition1 and is associated with several periparturient diseases and an increase in the calving-toconception interval. In dairy cows, fatty liver occurs primarily in the first 4 weeks after calving when up to 50% of all cows have some accumulation of triacylglycerol in the liver. A severe form of fatty infiltration of the liver immediately before or after parturition is known as the fat-mobilization syndrome, fat-cow syndrome, or pregnancy toxemia of cattle, and it can be highly fatal. In beef cattle, the disease occurs most commonly in late pregnancy when the nutrient intake is decreased in cattle that were previously well fed and in good body condition. In a field study, the percentage of cattle dying or being culled because of disease was affected by the amount of hepatic triglyceride: 15%, 31%, and 42% for cattle with mild, moderate, and severe hepatic lipidosis, respectively. Outbreaks of the disease have occurred in dairy herds in which up to 25% of all cows were affected, with a case-fatality rate of 90%. Cattle have been classified into three groups on the basis of liver fat content determined histologically 1 week after parturition. Less than 20% lipid corresponds to less than 50 mg/g liver by weight; 20% to 40% lipid, 50 to 100 mg/g liver; and greater than 40% represents more than 100 mg/g liver. These concentrations correspond to mild, moderate, and severe cases of fatty infiltration, respectively. Cows with less than 20% lipid in the liver at 1 week after calving are considered normal, and those with more than 20% are

1717

considered to have a fatty liver. About 30% of high-yielding dairy cows in the United Kingdom are considered to have a fatty liver 1 week after calving. Clinical evidence of hepatic disease may not occur consistently until liver lipid concentrations are in the range of 35% to 45% or more. Risk Factors Host Factors Fatty infiltration of the liver is part of a generalized fat-mobilization syndrome that occurs in early lactation, particularly in high-yielding dairy cows, as milk production outstrips appetite and body reserves are used to meet the energy deficit. In about 30% of high-producing cows, fatty infiltration in the liver is severe and is associated with reversible but significant effects on liver structure and function. In some populations of cows, the incidence of fatty liver is much lower and insignificant. Diseases that occur commonly in early lactation predispose to fatty liver include ketosis, left-side displacement of the abomasum, mastitis, retained fetal membranes, milk fever, and downer-cow syndrome. Any disease of early lactation that affects appetite and voluntary intake can contribute to fatty liver. The deficit occurs because dietary intake cannot meet the energy requirements for the high yield. Peak yields of milk are reached 4 to 7 weeks after calving, but the highest levels of voluntary feed intake are not reached until 8 to 10 weeks after calving. As a result of the energy deficit, the cow mobilizes body reserves for milk production and may lose a large amount of body weight. The BCS at calving can have a direct effect on the health, milk yield, and fertility of cows. It represents the cumulative effects of the dry period, the BCS at drying off, and the loss of body condition during the dry period. The risk of retained placenta may be greater for cows underconditioned at drying, whereas cows that lost more body condition during the dry period may be more affected by both retained placenta and metritis; the two effects are independent of each other. The risk of ketosis is increased in cows overconditioned at calving, which may be a result of a long dry period. Cows calving at a higher BCS produced more milk, fat, and protein in the first 90 days of lactation, and the effect was most pronounced for milk-fat content. Cows with a higher BCS at calving were less prone to anestrus, but they did not conceive more successfully to first service. A reduction of 6 open days in primiparous cows was estimated for each additional unit of BCS at calving. Multiparous cows that lose more body condition during the dry period are more prone to inactive ovaries and are more likely to be open 150 days after calving in the next lactation. Dairy cows with abnormally long dry periods also have a tendency to become

1718

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

obese and develop the fatty liver syndrome of parturition. The feeding of dairy cows in large groups, as in loose housing systems, has been associated with an increase in the incidence of the disease. The disease has occurred in pregnant heifers within 31 days after being turned out onto grass. The disease can occur in nonlactating dairy cows by the imposition of a partialstarvation diet in late pregnancy in an attempt to reduce the body weight of cows that are considered to be too fat. Changing the diet of pregnant beef cows from silage to straw in an attempt to reduce their body weight and the incidence of dystocia has resulted in outbreaks of the disease. In beef cattle in North America, the severe form of the disease, pregnancy toxemia, is seen most commonly in the last 6 weeks of pregnancy in cows that are fat and pregnant with twins. The affected cows are usually well fed until late pregnancy, when an unexpected shortage of feed occurs, or the cows are too fat and cannot consume sufficient low-energy feed to meet the demands of pregnancy. Under usual circumstances, the disease in beef cattle occurs sporadically: the morbidity is about 1%, but the mortality is usually 100%. Pregnancy toxemia of cattle has occurred in pregnant beef cattle in Australia and the United Kingdom. First-calf heifers were more commonly affected than older cows, and most were in late pregnancy (7 to 9 months) or had just recently calved. Cows pregnant with twins are particularly susceptible. Genetics of Lipid Mobilization Cows generally mobilize body lipid reserves in early lactation and regain these reserves during subsequent pregnancy. Lipid mobilized from body reserves makes a substantial contribution to the energetic cost of milk production in early lactation. It is usually assumed that this mobilization of body energy reserves is entirely a response to a deficit in feed energy intake relative to milk energy output. This implies that increasing the energy content of the feed being offered would decrease body energy mobilization in early lactation. A number of studies indicate that this is not always the case. It has been proposed that mobilization of body reserves in early lactation and the subsequent gain in body reserves during pregnancy are to a large extent genetically driven. Genetically driven bodylipid change is defined as that which would occur in cows kept in an environment that was in no way constraining. It then follows that environmentally driven body-lipid change is defined as that which occurs in response to an environment that is constraining. The rationale and evidence for genetically driven body-lipid change have their basis in evolutionary considerations and in the changes in lipid metabolism throughout the reproductive cycle.

Environmental and Dietary Factors In North America, the introduction of the system of challenge feeding of dairy cows was associated with an increased incidence of fatty liver. The overall effect of the system is to provide excess energy in the diet during late pregnancy or during the dry period generally. The diets fed may contain a high percentage of the cereal grains, corn ensilage, or brewer’s grains. In this system, high-energy rations are fed beginning a few weeks before parturition. The total daily amount of feed is increased by regular increments to reach a high level at parturition and peak levels to coincide with the peak in the lactation curve several weeks after parturition. This resulted in some excessively fat cows at the time of parturition, when energy demands are high. The disease has also occurred in dairy cows that were fed excessive amounts of high-energy rations throughout the dry period. In dairy herds, fatty liver syndrome has also been associated with an increase in the incidence of milk fever, ketosis, and leftsided displacement of the abomasum, all of which are much more difficult to treat successfully because of the fatty liver. Overfeeding during the dry period predisposes cows to accumulate fat in adipose tissue during the prepartum period. Before parturition, adipose tissue from overfed cows has higher rates of esterification than the adipose tissue of cows fed a restricted energy intake. In the fatty livers of these overfed cows, the rate of gluconeogenesis is not optimal, which results prolongation of lipolysis, particularly during the first few weeks after parturition. The increased lipolysis after parturition leads to a major increase in the hepatic triacylglycerol concentration and to a shift in hepatic fatty acid composition. Unrestricted feed intake during the dry period impairs postpartum oxidation and synthesis of fatty acids in the liver of dairy cows. In Australia, only beef cattle have been involved in pregnancy toxemia; the fat and the obese are most commonly affected. The disease occurred most notably when there was a shift to autumn calving (February to April) when feed supplies were low because of low late-summer rainfall. The cows were in good to fat body condition because of lush pastures in the spring and early summer, but by autumn when the calving season approached, the feed supplies were low and the nutritive value of the pasture inadequate. The lack of feed combined with the expensive nature of supplementary feeding resulted in an inadequate level of nutrition during late pregnancy. The morbidity is usually from 1% to 3%, but may be as high as 10%, and the disease is usually fatal.

PATHOGENESIS

Fatty liver is associated with a negative energy balance that is essentially universal in dairy cows in the first few weeks of lactation.

Most cows adapt to the negative energy balance through an intricate mechanism of metabolic adaptation. Fatty liver develops because of failure of these adaptive mechanisms. Under normal physiologic conditions, the total amount of fat increases in the liver beginning a few weeks before calving, rises to an average of about 20% (of wetweight basis) 1 week after calving, and declines slowly to the normal level of less than 5% by 26 weeks after calving. However, the fat content varies from almost none to 70% among cows 1 week after calving. Fat mobilization begins about 2 to 3 weeks before calving and is probably induced by a changing hormonal environment before calving rather than an energy deficit. After calving, there is a larger increase in fat accumulation. The changes in the liver in dairy cows are functional and reversible and related to the metabolic demands of late pregnancy and early lactation. The heavy demands for energy in the high-producing dairy cow immediately after parturition, or in the pregnant beef cow that may be bearing twins, result in an increased rate of mobilization of fat from body reserves, usually subcutaneous fat, to the blood that transports it to body tissues, particularly the liver but also muscle and the kidneys. Any decrease in energy intake caused by a shortage of feed or an inability of the cow to consume an adequate amount of feed during the critical periods of late pregnancy or early lactation results in the mobilization of an excessive amount of nonesterified fatty acids (NEFAs). This results in increased hepatic lipogenesis with accumulation of lipid in enlarged hepatocytes, depletion of liver glycogen, and inadequate transport of lipoprotein from the liver. Most of the lipid infiltration of the liver in dairy cows after calving is in the form of triacylglycerols because of the increased uptake of NEFAs and a simultaneous increase in diacylglycerol acyltransferase; the activity of this enzyme is activated by fatty acids. The gradual increase in plasma NEFA concentration during the final prepartum days may explain the gradual depression in dry matter intake and a contributing factor to triglyceride accumulation in the liver. During this period there is also an elevated concentration of plasma glucose and a lowered plasma BHB concentration. The serum lecithin : cholesterol acyltransferase activity in spontaneous cases of fatty liver in cows is also decreased, which may be associated with reproductive performance because cholesteryl esters are utilized for the synthesis of steroid hormones. Cattle are prone to fatty liver because their hepatocytes have limited capacity to export VLDLs and therefore a limited ability to export accumulated fat in the hepatocytes. NEFAs transported to the liver are usually oxidized in the mitochondria and peroxisomes or secreted as VLDL particles into the blood. Fatty liver develops when the

Metabolic Diseases of Ruminants

VetBooks.ir

uptake of NEFAs by the liver exceeds the oxidation of NEFAs by the liver to CO2, partial oxidation of NEFAs to form ketones, and export of phospholipids, cholesterol, and apoproteins from the liver as lipoproteins. For unknown reasons, the capacity for VLDL formation is low in cattle and further impaired in early-lactating cows a result of very low apoliprotein B100 (apoB100) availability, the main apolipoprotein of VLDL particles. Production of ketones in moderate levels is beneficial in that energy is exported from the liver to other tissues that can utilize ketones as an energy source. Excess lipids that cannot be exported are stored as triacylglycerol in the liver and are associated with decreased metabolic functions of the liver. Also, a prepartum surge of estrogen may contribute to the development of fatty liver in ruminants by increased fatty acid esterification along with limited export of triglyceride. During fat mobilization, there is a concurrent loss of body condition and adipose tissue. The degree of mobilization will be dependent on the fatness of the cow and extent of the energy deficit. Fat and thin cows respond differently to the metabolic demands of early lactation. Fat cows appear less able to utilize mobilized fatty acids, and as a result they accumulate esterified fat in tissues. This can adversely influence susceptibility to disease, and the response of the cow to that disease imposes further metabolic demands, particularly on muscle and protein metabolism. Both fat and skeletal muscle mass are decreased after calving, and fat cows lose 2.5 times more muscle fiber area than thin cows. Thus the loss of body condition is a result of total tissue mobilization (protein and fat) rather than fat alone. There appears to be a higher rate of protein mobilization in fat cows than in thin cows. Cows that are not fat initially do not develop fatty liver syndrome. Pregnant beef cows in thin body condition on pasture can become extremely emaciated and eventually recumbent and die of starvation, but they do not develop pregnancy toxemia.

CLINICAL FINDINGS

In dairy cattle, fat-cow syndrome occurs usually within the first few days following parturition and is commonly precipitated by any condition that interferes with the animal’s appetite temporarily, such as the following: • Parturient hypocalcemia • Left-sided displacement of the abomasum • Indigestion • Retained fetal membranes • Dystocia Affected cows are usually excessively fat, with a BCS of 4/5 or higher. Excessive quantities of subcutaneous fat are palpable over the flanks, the shoulder areas, and around the tailhead. The affected cow usually does

not respond to treatment for some of these diseases and becomes anorexic. The temperature, heart rate, and respiration are within normal ranges. Rumen contractions are weak or absent, and the feces are usually scant. Periods of prolonged recumbency are common, and affected cows may have difficulty in standing when they are coaxed to stand. A severe ketosis that does not respond to the usual treatment may occur. There is marked ketonuria. Affected cows will not eat and gradually become weaker and progress to totally recumbent, and they die in 7 to 10 days. Some cattle exhibit nervous signs consisting of a staring gaze, holding the head high, and muscular tremors of the head and neck. Some severe cases appear to develop hepatic failure, do not respond to therapy, and become weak and recumbent and die. Terminally there is coma, tachycardia, and marked hyperglycemia. The case-fatality rate in severe cases may reach 50% or more. In fat beef cattle shortly before calving, affected cows are aggressive, restless, excited, and uncoordinated with a stumbling gait; sometimes have difficulty in rising; and they fall easily. The feces are scant and firm, and there is tachycardia. When the disease occurs 2 months before calving, the cows are depressed for 10 to 14 days and do not eat. Eventually they become sternally recumbent. The respirations are rapid, there may be an expiratory grunt, and the nasal discharge is clear, but there may be flaking of the epithelium of the muzzle. The feces are usually scant; terminally, there is often a fetid yellow diarrhea. The disease is highly fatal; the course is 10 to 14 days, and terminally there may be coma, with cows dying quietly. In dairy cattle with moderately severe fatty liver, the clinical findings are much less severe, and most will recover within several days if they continue to eat even small amounts of hay. In dairy cattle, there is a relationship between the occurrence of a subclinical fatty liver within the first few weeks after parturition and inferior reproductive performance as a result of a delay in the onset of normal estrus cycles and a reduction in the conception rate that results in an increase in the average days between calving and conception. There may be differences in reproductive performance between cows with mild and moderate fatty livers early after calving. However, an examination of the postpartum hormone profiles of cows with fatty liver did not reveal the pathogenic mechanism of the reduced fertility. Fat-cow syndrome may also be associated with an increased incidence of parturient paresis and unresponsive treatment for ketosis in early lactation.

CLINICAL PATHOLOGY Serum Biochemistry The biochemical changes associated with fatty liver syndrome in cows depend on the severity of the fatty liver. There is a

1719

significant association between increasing serum biochemical abnormalities with increasing amounts of liver fat, although there may be considerable overlap in the distribution of individual test values in a population of animals with suspected fatty liver. Increased plasma/serum nonesterified fatty acid, acetoacetate, BHB, and total bilirubin concentrations, and decreased serum fructosamine concentration,2 are associated with increased liver fat percentage. Likewise, increased plasma/serum hepatic enzyme activity (particularly aspartate aminotransferase and ornithine carbamoyl transferase activity) is also associated with increased liver fat percentage. Other hepatic enzyme activities in plasma, such as alanine aminotransferase, sorbitol dehydrogenase, glutamate dehydrogenase, alkaline phosphatase, and gamma-glutamyl transferase activities, are poorly associated with liver fat percentage.1 Possibly the most relevant biochemical index of the liver fat percentage is the plasma NEFA : cholesterol ratio. The rationale for using this ratio is that the plasma NEFA concentration reflects a metabolite that has not been cleared by the liver, whereas the plasma cholesterol concentration reflects the rate of hepatic reesterification and export as a VLDL. A high plasma NEFA : cholesterol concentration therefore is thought to indicate a high liver fat percentage. An increased concentration of plasma total bilirubin is also associated with increased liver fat percentage; competition between bilirubin and NEFA for the same binding site on hepatocytes decreases the hepatic uptake of bilirubin and therefore results in hyperbilirubinemia.2 Serum fructosamine concentration provides a retrospective record of serum/plasma glucose concentrations over the previous 1 to 3 weeks and therefore provides a useful longer-term index of glucose availability. Serum fructosamine concentrations less than 213 µmol/L are predictive of hepatic lipidosis in dairy cattle.2 The plasma ammonia concentration in arterial or venous samples is poorly associated with liver fat percentage, but it is an excellent indicator of hepatic failure in severely affected cattle with hepatic lipidosis.3 In cattle, ammonia in plasma is derived mainly from bacterial activity in the rumen and metabolism of tissue amino acids and is converted to urea by the liver or glutamine by the liver and other tissues. Consequently, severe liver dysfunction results in elevated plasma ammonia concentrations (>29 µmol/L), with higher ammonia concentrations in arterial samples than venous samples because of nonhepatic metabolism of ammonia. Several cow-side blood, urine, and milk ketone tests are available for the detection of subclinical ketosis in postpartum dairy cows (see previous section on ketosis and subclinical ketosis). Metabolomic biomarkers show promise in identifying a typically pattern

1720

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

of changes in cattle with hepatic lipidosis; for example, plasma fibrinogen decreases inversely with the severity of hepatic lipidosis, presumably because of intracellular lipid accumulation interferes with fibrinogen synthesis.4

affected and might have some clinical evidence of hepatic insufficiency. Those between 25% and 13% are mildly affected, which is the range of most postpartum dairy cows without any evidence of disease. Liver lipid concentrations below 13% are inconsequential.

Hemogram In cattle with subclinical fatty liver, there may be a leukopenia, neutropenia, and lymphopenia. Leukopenia has been observed in dairy cows with more than 20% liver fat in the second week after calving. This may be related to the increased incidence of postparturient diseases, such as mastitis and endometritis, observed in cows with subclinical fatty liver. In cows with fatty liver, there is decreased functional capacity of the polymorphonuclear cells. However, this is not necessarily a cause-and-effect relationship.

Ultrasonography of the Liver Ultrasonography of the liver has been used to evaluate fatty infiltration in dairy cattle with mixed results.5,6 Two strategies have been employed: identification of hepatic enlargement by comparing liver position with published reference range relative to the ribs, and the echogenicity or brightness of the liver. In the normal cow, the hepatic ultrasonogram consists of numerous weak echoes distributed homogeneously over the entire area of the liver. The echo beam gradually attenuates as it passes through the normal liver tissue. The portal and hepatic veins can be seen within the normal echotexture, and the parenchymal edges are normally visible. In the fatty liver, there is a diffuse nature and echogenicity that are roughly proportional to the volume of fat vacuoles and the amount of triglyceride in the liver. Assessment of echogenicity is subjective and varies with equipment and settings on the ultrasonographic unit. Consequently, objective ultrasonographic indices of hepatic lipidosis are under investigation, such as spectral analysis and analysis of brightness (B)-mode image statistics and texture characteristics.6 Technological challenges associated with digital processing of ultrasonographic images of the liver need to be resolved before the noninvasive measurement of liver fat percentage becomes a widely available diagnostic tool.

Liver Biopsy and Analysis The severity of fatty liver has been arbitrarily classified into severe, moderate, and mild, based on the amount of triglyceride present in the hepatocytes.1 In severe hepatic lipidosis, the accumulation of triglyceride in the cytoplasm is accompanied by disturbances in hepatic structure and function that may result in hypoglycemia and ketonemia; these signs are manifested as anorexia and depression, and there may be clinical evidence of nervous signs. A liver biopsy can be used to determine the severity of the fatty liver and the concentration of triglyceride and is the most reliable method of accurately estimating the degree of fatty infiltration of the liver. The triglyceride concentration of liver in normal cows ranges from 10% to 15% on a wet-weight basis. Estimation of the lipid content of bovine liver samples obtained by biopsy may be made by biochemical or histologic methods. Both methods provide reasonable estimates of liver fat content over a wide range of values. The lipid content of bovine liver is highly correlated with its specific gravity and the submersion of needle biopsy specimens into water, and copper sulfate solutions with specific gravities of 1.025 and 1.055 can be used as a test to estimate lipid content. For routine clinical diagnosis, three solutions of specific gravities of 1, 1.025, and 1.055 can be used. Liver samples that float in all three solutions contain greater than 34% lipid, those that sink in water but float in solutions of 1.025 and 1.055 specific gravity contain less than 34% but greater than 25% lipid, whereas those that float only in solutions of 1.055 specific gravity contain less than 25% but greater than 13% lipid. Samples that sink in all three solutions contain less than 13% lipid. Some limited evidence indicates that cows with liver lipid concentrations above 34% are severely affected and can be expected to have clinical manifestations of hepatic insufficiency. Those with liver lipid levels between 34% and 25% are moderately

NECROPSY FINDINGS

In severe fatal cases, the liver is grossly enlarged, pale yellow, friable, and greasy. Mild and moderate cases are usually not fatal unless accompanied by another fatal disease, such as peracute mastitis. The degree of fatty infiltration in these instances is much less obvious. The histologic changes include the occurrence of fatty cysts or lipogranulomas, enlarged hepatocytes, compression of hepatic sinusoids, a decreased volume of rough endoplasmic reticulum, and evidence of mitochondrial damage. The latter two changes are reflected in reduced albumin levels and increased activities of liver enzymes in the blood. The proportions of the various fatty acids in the liver are altered considerably. Palmitic and oleic acid proportions are higher in fatty-liver cows than in normal cows, whereas stearic acid is lower. DIFFERENTIAL DIAGNOSIS In dairy cows, fatty liver must be differentiated from those diseases that occur commonly immediately following parturition. Left-sided displacement of the abomasum

results in a secondary ketosis, inappetence, and pings over the left abdomen. Retained placenta and metritis may be accompanied by fever, inappetence to anorexia, ruminal atony, and a foul-smelling vaginal discharge. A degree of fatty liver may occur in these cows, making it indistinguishable from the effects of the retained placenta and metritis. Primary ketosis may occur immediately after parturition or within several days rather than at the most common time, at 6 to 8 weeks of lactation. Inappetence, ruminal hypotonicity, marked ketonuria, and a good response to glucose and propylene glycol are characteristic. In beef cattle, pregnancy toxemia before parturition must be differentiated from abomasal impaction, vagus indigestion, and chronic peritonitis.

TREATMENT The prognosis for severe fatty liver is unfavorable. In general, cows with the severe fat-cow syndrome that are totally anorexic for 3 days or more usually die in spite of intensive therapy. The prognosis for cases with nervous signs is very poor. Liberal quantities of highly palatable good-quality hay and an ample supply of water should be provided. Cattle that continue to eat in increasing daily amounts will recover with supportive therapy and palatable feeds. The major prognostic factor is whether the cow will eat; failure of the appetite to return is usually a very poor prognostic sign. Three treatment strategies for fatty liver are available. The most effective strategy is to decrease the rate of fat mobilization and therefore the plasma NEFA concentration; propylene glycol appears to act partly by this mechanism. The second strategy is to facilitate the complete oxidation of NEFAs in the liver. The third strategy is to increase the rate of export of VLDLs from the liver; choline is thought to act by this method. Because they address different mechanisms, combined treatment using propylene glycol and rumen-protected choline offers theoretical advantages. Several different therapeutic approaches have been tried and are discussed in detail in the previous section on ketosis and subclinical ketosis Additional treatments that have been tried in cattle with fatty liver include intravenous fluids, ruminal transfaunation, and glucagon. Fluid and Electrolyte Therapy.  Intensive therapy directed at correcting the effects of the ketosis and the fatty liver is required. The recommended treatment includes continuous IV infusion of 5% glucose and multiple electrolyte solutions and the intraruminal administration of rumen juice (5 to 10 L) from normal cows in an attempt to stimulate

Metabolic Diseases of Ruminants

VetBooks.ir

the appetite of affected cows. Water and multiple electrolytes (10 to 30 L) can be administered intraruminally. Glucagon.  The subcutaneous injection of 15  mg/d of glucagon for 14 days beginning at day 8 postpartum decreases liver triglyceride concentrations in cows older than 3.5 years. Glucagon, containing 29 amino acids, is a pancreatic hormone that improves the carbohydrate status of cows by stimulating hepatic gluconeogenesis, glycogenolysis, amino acid uptake, and ureagenesis. The effect of glucagon on lipid metabolism is both direct and indirect because it directly increases lipolysis in adipose tissue but indirectly decreases lipolysis by increasing concentrations of plasma glucose and insulin. IV infusions of glucagon are not practical for on-farm use. Glucocorticoids.  Dexamethasone-21isonicotinate (20 to 25 mg, IM) decreases hepatic total lipid and triglyceride content in cattle after surgical correction of leftdisplaced abomasum, which is a beneficial effect.7 Propylene glycol given orally at 300 mL/ day for 5 days promotes gluconeogenesis and is used for the treatment of ketosis. Insulin as zinc protamine at 200 to 300 SC twice daily promotes the peripheral utilization of glucose, but clinical results have been mixed. It is important to recognize that IV administration of glucose is always accompanied by insulin release, no matter the metabolic state of the cow. Consequently, IV glucose administration should be considered as a combined treatment with glucose and insulin. Outbreaks in a Herd.  When outbreaks of fat-cow syndrome occur in pregnant beef cattle, all remaining cows should be sorted into groups according to body condition and fed accordingly. Excessively fat cows should be fed the best-quality hay that is available along with a supplement. Fat cows should be exercised by feeding them on the ground and forcing them to walk.

TREATMENT AND CONTROL Treatment Propylene glycol (300 mL daily for 5 days, PO) (R-1) Dextrose (500 mL of 50% dextrose once, IV) (R-1) Dexamethasone, dexamethasone-21isonicotinate, or flumethasone, IM (R-1) Cyanocobalamin (vitamin B12, 1 to 4 mg IV, daily for 2 to 3 treatments) (R-2) Isoflupredone (20 mg, IM, multiple injections) (R-3)

Control Monensin (controlled-release capsule, 335 mg/ day) (R-1) Propylene glycol (300 to 500 mL daily for 5 days, PO) (R-1) Cyanocobalamin (vitamin B12, 1 to 4 mg IV, daily for 2 to 6 treatments before or at calving) (R-2)

CONTROL Control and prevention of fatty liver in cattle will depend on decreasing or eliminating most of the potential risk factors for the disease. Early recognition and treatment of diseases that affect the voluntary dietary intake in late pregnancy and immediately after parturition are necessary to minimize the mobilization of body-fat stores to meet the overall energetic requirements of the cow during the period of negative energy balance and to maintain or increase hepatic glucogenesis. Diseases such as ketosis, displaced abomasum, retained placenta, acute mastitis, milk fever, and downer-cow syndrome must be treated as early as possible to avoid varying degrees of hepatic lipidosis. Dry Matter Intake and Energy Balance in the Transition Period The literature on dry matter intake and energy balance in the transition period of the dairy cow has been reviewed. The transition from late gestation to early lactation in the dairy cow is a critical period in the lactation–gestation cycle. During this period, feed intake is at the lowest level in the production cycle. In addition to the drop in feed intake, there is a concurrent transition from late gestation to lactation, with huge increases in energy demands. This leads to a negative energy balance that can result in ketosis or fatty liver. Voluntary dry matter intake (DMI) may decrease 25% and 52% during the final 14 days of gestation for first- and secondparity animals and aged (third and fourth or greater) cows, respectively. A negative energy balance can occur before parturition and is more likely to occur in heifers than cows because heifers have a lower DMI and an additional need for energy requirement for growth. The fall in DMI is the usual cause of a negative energy balance rather than an increase in energy requirements for fetal growth. Metabolic Adaptations During the Transition Period The primary goal of nutritional management strategies of dairy cows during the transition period should be to support the metabolic adaptations that occur. The hallmark of the transition period of dairy cattle is the dramatic change in nutrient demands that necessitates exquisite coordination of

1721

metabolism to meet requirements for energy, amino acids, and calcium by the mammary gland after calving. Estimates of the demand for glucose, amino acids, fatty acids, and net energy by the gravid uterus at 250 days of gestation and the lactating mammary gland at 4 days postpartum indicate approximately a tripling of demand for glucose, a doubling of demand for amino acids, and approximately a fivefold increase in demand for fatty acids during this period. In addition, the requirement for calcium increases approximately fourfold on the day of parturition. The literature on the integration of metabolism and intake regulation in periparturient animals has been reviewed. Glucose Metabolism The primary homeorhetic adaptation of glucose metabolism to lactation is the concurrent increase in hepatic gluconeogenesis and decrease in oxidation of glucose by peripheral tissues to direct glucose to the mammary gland for lactose synthesis. The major substrates for hepatic gluconeogenesis are propionate from ruminal fermentation, lactate from Cori cycling, amino acids from protein catabolism or net portal-drained visceral absorption, and glycerol released during lipolysis in adipose tissue. Lipid Metabolism The primary homeorhetic adaptation of lipid metabolism to lactation is the mobilization of body fat stores to meet the overall energetic requirements of the cow during a period of negative energy balance in early lactation. Body fat is mobilized into the bloodstream in the form of NEFAs that are used to make upward of 40% of milk fat during the first days of lactation. Skeletal muscle uses some NEFA for fuel, particularly as it decreases its reliance on glucose as a fuel during early lactation. Given that NEFA concentrations increase in response to increased energy needs accompanied by inadequate feed intake, and plasma NEFA concentrations usually are inversely related. The liver takes up NEFAs in proportion to their supply, but the liver typically does not have sufficient capacity to completely dispose of NEFAs through export into blood or catabolism for energy. Therefore cows are predisposed to accumulate NEFAs as triglycerides within liver when large amounts of NEFA are released from adipose tissue into the circulation. Nutritional Management to Support Metabolic Adaptations During the Transition Period Grouping Strategies The primary goal of nutritional management strategies of dairy cows during the transition period should be to support the metabolic adaptations just described. Industrystandard nutritional management of dairy cows during the dry period consists of a two-group nutritional scheme. The National

1722

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Research Council (NRC) Nutrient Requirements of Dairy Cattle recommends that a diet containing approximately 1.25 Mcal/kg of NEL should be fed from dry-off until approximately 21 days before calving, and that a diet containing 1.54 to 1.62 Mcal/kg of NEL should be fed during the last 3 weeks before calving. The primary rationale for feeding a lower-energy diet during the early part of the dry period is to minimize BCS gain during the dry period. During the last 3 to 4 weeks prepartum, a diet higher in energy and protein concentration than current NRC recommendations should be fed so that adequate nutrient intake occurs within the limits of the reduced voluntary dry matter intake. Supplying excessive energy to dairy cows during the early dry period may have detrimental carryover effects during the subsequent early lactation. Managing cows to achieve a BCS of approximately 3.0 at drying off rather than the traditional 3.5 is now recommended. Strategies to Meet Glucose Demands and Decrease NEFA Supply During the Transition Period Carbohydrate Formulation of the Prepartum Diet.  Feeding diets containing higher proportions of nonfiber carbohydrate (NCF) promotes ruminal microbial adaptation to NFC levels typical of diets fed during lactation and provides increased amounts of propionate to support hepatic gluconeogenesis and microbial protein (providing the diet contains sufficient ruminally degradable protein) to support protein requirements for maintenance, pregnancy, and mammogenesis. Direct Supplementation With Glucogenic Precursors.  Propylene glycol is a glucogenic precursor that has been used as an oral drench in the treatment of ketosis. Decreased concentrations of plasma NEFA and BHB follow oral administration of propylene glycol. The administration of an oral drench of propylene glycol for 2 days beginning at calving decreased concentrations of NEFA in plasma and increased milk yield during early lactation. However, in general, the lack of consistent production responses does not support a recommendation for routine use. Propionate supplements added to the diet to supply substrate for hepatic gluconeogenesis have also been used, but with inconsistent results. Glycerol given orally is an effective treatment for lactational ketosis in dairy cattle. Feeding glycerol to dairy cows from 14 days prepartum to 21 days in milk did not have the glucogenic effect attributed to it when given orally as a drench to individual cows. Monensin provided in controlled-release capsules (CRCs) administered 2 to 4 weeks prepartum has been shown to decrease the incidence of energy-associated diseases, subclinical ketosis, and left-side displaced abomasum by 40%, and a 25% reduction in

retained placenta was found. The capsule delivers 335 mg/d of monensin for 95 days. The common mechanism for reduction of the incidences of these energy-associated diseases is likely to be improved energy metabolism during the transition period. The net effect of monensin within the rumen is to increase ruminal propionate production at the expense of ruminal acetate and methane production so that propionate supply is increased and the overall energetic efficiency of ruminal fermentation is increased. Added Fat in Transition Diets.  It has been proposed that dietary fat may partially decrease concentrations of NEFA and prevent the occurrence of ketosis. Dietary long-chain fatty acids are absorbed into the lymphatic system and do not pass first through the liver. The fat can provide energy for peripheral tissues and the mammary gland, and the increased energy availability would in turn decrease mobilization of body fat and decrease plasma NEFA concentrations. However, available evidence indicates that added fat fed to cows during the prepartum period does not decrease plasma NEFA concentrations. Effects of Specific Fatty Acids on NEFA Supply.  A substantial amount of research has examined the metabolic roles of individual fatty acids in transition-cow nutrition and metabolism. Feeding trans-10, cis-12 conjugated linoleic acid or transoctadecanoic acid experimentally may decrease the negative energy balance, but the ultimate metabolic effects in transition cows are as yet uncertain. Because of the large economic losses associated with pregnancy toxemia in cattle, every economic effort must be made to prevent the disease. The principal method of control is to prevent pregnant cattle from becoming fat during the last trimester of pregnancy, particularly during the dry period in dairy cattle. During pregnancy, mature cattle should receive sufficient feed to meet the needs for maintenance and pregnancy, and the total daily nutrient intake must increase throughout the last trimester to meet the needs of the fetus. However, this increase is usually difficult to control without some cows getting fat and others losing weight. Sorting cows into groups on the basis of size and condition and feeding accordingly is recommended. Metabolic profiles may be used as a means of assessing energy status and, correspondingly, the likelihood of occurrence hyperketonemia or pregnancy toxemia. Both plasma glucose and BHB concentrations can be used. Body-condition scoring of dairy cows at strategic times can be used to monitor the nutritional status of the herd and minimize the incidence and severity of fatty liver syndrome. The scoring should be done throughout the production cycle as part of a herd

health program. Scoring done at calving, at 21 to 40 days, and 90 to 110 days postpartum can be used to monitor the nutritional status of the herd. Scoring done at 100 to 60 days before drying off provides an opportunity for management to make appropriate adjustments in the feeding program so that optimal body-condition goals are achieved. The optimum BCS of a cow at calving that will result in the most economical amount of milk has not yet been determined. On a scale of 5, the suggested optimum score at calving has ranged from 3 to 4. The optimum score will probably depend on the characteristics of the individual herd, which include type of cow, type of feedstuffs available, season of the year, environmental temperature, and the people doing the actual body-condition scoring. FURTHER READING Gordon JL, LeBlanc SJ, Duffield TF. Ketosis treatment in lactating dairy cattle. Vet Clin North Am Food Anim Pract. 2013;29:433-445. Grummer RR. Nutritional and management strategies for the prevention of fatty liver in dairy cattle. Vet J. 2008;176:10-20. Ingvartsen KL. Feeding and management related diseases in the transition cow. Physiological adaptations around calving and strategies to reduce feeding-related diseases. Anim Feed Sci Technol. 2006;126:175-213. Ringseis R, Gessner CK, Eder K. Molecular insights into the mechanisms of liver-associated diseases in early-lactating dairy cows: hypothetical role of endoplasmic reticulum stress. J Anim Physiol Anim Nutr. 2015;99:626-645.

REFERENCES

1. Kalaitzakis E, et al. J Vet Intern Med. 2007;21:835. 2. Mostafavi M, et al. Anim Prod Sci. 2015;55:1005. 3. Mudron P, et al. Vet Med Czech. 2004;49:187. 4. Imhasly S, et al. BMC Vet Res. 2014;10:122. 5. Rafia S, et al. Am J Vet Res. 2012;73:830. 6. Starke A, et al. J Dairy Sci. 2010;93:2952. 7. Kusenda M, et al. J Vet Intern Med. 2013;27:200.

PREGNANCY TOXEMIA (TWIN LAMB DISEASE) IN SHEEP SYNOPSIS Etiology A multifactorial disorder of energy metabolism, with hypoglycemia and ketonemia (the accumulation in blood of acetoacetate, β-hydroxybutyrate, and their decarboxylation products acetone and isopropanol). Epidemiology The disease in sheep is associated with a falling plane of nutrition, principally in the last month of pregnancy in ewes bearing twins and triplets, but can be induced by other stress at this time. Clinical findings Encephalopathy with blindness, muscle tremor, convulsions, metabolic acidosis, and a clinical course of 2 to 8 days, usually terminating fatally unless treated early.

Metabolic Diseases of Ruminants

VetBooks.ir

Clinical pathology Hypoglycemia, ketonemia, ketonuria. Necropsy findings None specific. Twin lambs and fatty liver. Diagnostic confirmation Ketonemia, ketonuria, or elevated ketones in milk. Elevated β-hydroxybutyrate (BHBA) in aqueous humor of dead sheep. Treatment Parenteral glucose with corticosteroid and oral glucose precursors such as propylene glycol, occasionally insulin, or oral glucose and electrolyte therapy. Cesarean section or induction of parturition. Case fatality high. Control Monitoring of condition score, pasture availability, feeding, and biochemical indicators of ketosis. Correction of energy imbalance if detected.

ETIOLOGY Hypoglycemia and hyperketonemia are the primary metabolic disturbances in pregnancy toxemia. The precipitating cause is the energy demand of the conceptus in the latter part of pregnancy. However, there is a great deal of variation between sheep flocks in the prevalence of the naturally occurring disease under conditions that appear conducive to its development. The most important factor in pregnancy toxemia is a decline in the plane of nutrition during the last 4 to 6 weeks of pregnancy. This is the period when fetal growth is rapid and the demands for energy are markedly increased, particularly in ewes carrying twins or triplets. For example, the energy requirement for a 70-kg ewe carrying twins increases 36% in the last weeks of pregnancy, from 13.5 MJ (3.2 Mcal)/d midgestation to 18.3 MJ (4.4 Mcal)/d at term. The disease in goats during late pregnancy has the same initiating causes. Pregnancy toxemia can be classified according to the underlying management cause that is critical to its control and prevention: • Primary pregnancy toxemia • Fat-ewe pregnancy toxemia • Starvation pregnancy toxemia • Secondary pregnancy toxemia • Stress-induced pregnancy toxemia

EPIDEMIOLOGY Primary Pregnancy Toxemia Primary pregnancy toxemia is the most common. In most flocks it is a result of a declining plane of nutrition in the latter half of pregnancy, often exacerbated by a short period of food deprivation associated with a management procedure in late pregnancy, such as crutching, shearing, a change of environment, or drenching. In sheep grazing pastures the decreased plane of nutrition is often associated with inadequate pasture availability and/or overstocking. In sheep at pasture it occurs more frequently in

early-lambing flocks, where there is insufficient supplement provided during autumn or winter. In some outbreaks ewes have been moved onto better pasture during late pregnancy specifically to prevent the occurrence of ketosis, but if ewes are unaccustomed to the new feed, their intake of metabolizable energy will be reduced. For sheep that are housed in late pregnancy, the provision of poor-quality hay may predispose pregnancy toxemia. A change in feed type, feeding of moldy feed, or feed contaminated with manure can also lead to decreased intake, especially with goats. Competition for inadequate trough space can also be important. Goats exhibit greater dominant/submissive behavior than sheep, and this can result in lower food intake in submissive goats in groups that are being fed a partial supplement or total ration. In all management systems, failing to identify and separate ewes bearing twins and triplets, and to feed them accordingly, or failing to increase the nutritional plane of mixed mobs of pregnant sheep during the last 6 weeks of pregnancy are important predisposing factors. Fat-Ewe Pregnancy Toxemia Fat-ewe pregnancy toxemia occurs without a specific stressor in ewes that are very well fed and are in an overfat condition in late pregnancy (a condition score of 4 or 5 on a scale of 1 [emaciated] to 5 [fat]). Fat ewes have a decreased food intake in late pregnancy when the volume of the rumen is reduced by the pressure of intraabdominal fat and the developing fetus. This can occur especially when feeds with high water content are being fed, such as silage or root crops. A lack of exercise is thought to predispose this type of pregnancy toxemia, and there is often concurrent hypocalcemia. Starvation Pregnancy Toxemia Starvation pregnancy toxemia occurs in ewes that are excessively thin. It is relatively uncommon, but it occurs in extensive grazing systems where there is prolonged drought and an inadequate alternative feed supply. It can occur in any production system where there is mismanagement and undernutrition. Secondary Pregnancy Toxemia Secondary pregnancy toxemia usually occurs as a sporadic disease as the result of the effect of an intercurrent disease, such as foot rot or foot abscess, that affects food intake. Heavy worm infestation, such as mixed infections of Teladorsagia, Haemonchus, or Trichostrongylus species, would add a similar drain on glucose metabolism and increase the chances of development of this condition. Stress-Induced Pregnancy Toxemia Stress-induced pregnancy toxemia is the least common variant of this condition, in

1723

which stress is the initiator. Examples are the close shepherding or housing of latepregnant sheep of breeds not used to being housed, the transport of late pregnant sheep, and outbreaks following attack by dogs. Occurrence Pregnancy toxemia is seen primarily in ewes carrying triplet or twin lambs in the last 6 weeks of pregnancy, with the peak incidence in the last 2 weeks of pregnancy. It occurs wherever sheep are raised, but it is primarily a disease of sheep raised in intensive farming systems, either grazing or when housed during the winter. In part this is because the breeds of sheep used in intensive farming a re more likely to bear twins or triplets. In contrast, sheep breeds in extensive grazing systems commonly bear single lambs, and significant outbreaks of pregnancy toxemia are uncommon except where there is drought or insufficient pasture as a result of poor management. The attack rate in a flock varies with the nature and severity of the nutritional deprivation and the proportion of the flock at risk. It can be very high in starvation pregnancy toxemia, whereas fat-ewe pregnancy toxemia is generally sporadic. In outbreaks that follow management procedures or other stressors, clinical disease is not seen until 48 hours afterward, and new cases will develop over several days. Intercurrent disease in late-pregnant ewes, such as foot rot or foot abscess, may predispose pregnancy toxemia. The natural incidence in intensively farmed sheep is approximately 2% of pregnant ewes, but where there are severe management deficiencies it may affect the majority of late-pregnant ewes. The proportion of flocks with cases varies by year, but in a study of sheep diseases in Canada, 19% of flocks reported cases of pregnancy toxemia. The case fatality is high unless treatment is initiated early in the clinical course, but even with early treatment many ewes will die. Experimental Reproduction Hypoglycemia and ketosis can be experimentally produced in pregnant sheep by undernourishment, but the resultant syndrome has biochemical and clinical differences from spontaneously occurring pregnancy toxemia. For example, loss of appetite is an early sign in the spontaneous disease, whereas starved experimental animals, even though hypoglycemic and ketotic, will eat feed when offered. Consequently, there is debate about whether hypoglycemia is the primary precipitating cause of the clinical signs in the naturally occurring disease. There is a great deal of variation between sheep in the ease with which the hypoglycemia and ketosis can be produced experimentally and in the variation in incidence of the naturally occurring disease in conditions that appear to be conducive to it developing. It is likely that the difference between sheep

1724

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

depends on the metabolic efficiency of the liver. Animal Risk Factors Pregnancy The disease occurs only in ewes in the last 6 weeks of pregnancy, with the peak incidence in the last 2 weeks. It occurs primarily in ewes carrying triplet or twin lambs, although ewes bearing a single, large lamb may also be affected. Parity The disease is uncommon in maiden ewes because of their lower fecundity, and then increases in prevalence up until 5 to 6 years of age. Breed Breed differences largely reflect differences in fecundity and differences in management systems. Thus the disease is more common in British lowland breeds and their crosses than the Merino. British hill-breeds are traditionally thought to be more resistant to the development of pregnancy toxemia in the face of nutritional deprivation of the ewe, but resistance is achieved at the expense of lamb birth weight and has the penalty of higher neonatal mortality. Differences in the susceptibility of individual sheep appear to be related to differences in rates of hepatic gluconeogenesis. Economic Significance The disease has considerable effect. Without treatment, the case-fatality rate can approach 100%, and in individual flocks the prevalence can be high enough to be classed as an outbreak. Treated ewes that recover may have dystocia and die during parturition or develop retained placenta and metritis. Flocks that experience pregnancy toxemia also have a significantly higher-than-normal mortality in neonatal lambs and often a severe decrease in wool quality. Often these flocks are also predisposed to hypomagnesemia during lactation.

PATHOGENESIS

Approximately 60% of fetal growth takes place in the last 6 weeks of pregnancy, and pregnancy toxemia results from inadequate energy intake during this time, usually in ewes with more than one fetus. Ewes that are predisposed to the disease have an ineffective gluconeogenic response to the continued preferential demands for glucose by the growing fetuses, resulting in hypoglycemia, lipid mobilization, and the accumulation of ketone bodies and cortisol. The reason for this predisposition is not precisely known, but the subsequent disease and metabolic changes are associated with excessive lipid mobilization. Elevated concentrations of BHB further suppress endogenous glucose production and exaggerate the development of ketosis. Thus the negative feedback of

hyperketonemia on glucose production produces a self-perpetuating cycle. An encephalopathy develops, thought to be a hypoglycemic encephalopathy from hypoglycemia in the early stages of the disease. The encephalopathy and the disease are frequently not reversible unless treated in the early stages. The onset of clinical signs is always preceded by hypoglycemia and hyperketonemia, although it is not related to the minimum blood glucose or maximum ketone levels, and thus hypoglycemia may not be the initial or precipitating cause of the syndrome. In affected ewes, there is an abnormally high level of cortisol in plasma, and adrenal steroid diabetes (“insulin resistance”) may either contribute to or be a predisposing factor. For example, a comparison of ewes with a high risk of pregnancy toxemia (German Blackheaded Mutton) with a breed of lower risk (Finnish Landrace) found that the glucose elimination rate and glucose stimulated first-phase insulin secretion was lower and the basal rate of lipolysis significantly higher in the high-risk ewes. However, further investigation of insulin resistance and impaired insulin sensitivity, and the underlying cause of pregnancy toxemia, is needed.1 The increase in plasma concentrations of nonesterified fatty acids depresses cellular and humoral immune responses in the experimentally produced disease, but the clinical significance of this to naturally occurring disease is not clear.2 Renal dysfunction is also apparent in the terminal stages of ovine ketosis and contributes to the development of clinical signs and the fatal outcome. Those ewes that are carrying only one lamb and have been well fed before a short period of undernutrition may develop a subacute syndrome, both clinically and biochemically. In lines of ewe selected for increased fecundity, ewes bearing more than three fetuses have an increased susceptibility to pregnancy toxemia.3

CLINICAL FINDINGS

The earliest signs of ovine ketosis are separation from the group, altered mental state, and apparent blindness, manifested by an alert bearing but a disinclination to move. Sheep at pasture may fail to come up for supplementary feeding, and housed sheep may stand near the feed trough with other sheep but not eat. The ewe will stand still when approached by attendants or dogs and will turn and face them, but it will make no attempt to escape. If it is forced to move, it blunders into objects; when an obstacle is encountered, it presses against it with its head. Many affected ewes stand in water troughs all day and lap the water. Constipation with dry, scanty feces is common, and there is grinding of the teeth. In later stages, marked drowsiness develops, and episodes of more severe nervous signs occur, but they may be infrequent and easily missed. In these episodes, tremors of

the muscles of the head cause twitching of the lips, champing of the jaws, and salivation, and these are accompanied by a cog-wheel type of clonic contraction of the cervical muscles causing dorsiflexion or lateral deviation of the head, followed by circling. The muscle tremor usually spreads to involve the whole body, and the ewe falls with tonicclonic convulsions. The ewe lies quietly after each convulsion and rises normally afterward, but is still blind. Between the convulsions there is marked drowsiness that may be accompanied by head pressing; assumption of abnormal postures, including unusual positions of the limbs and elevation of the chin (the “stargazing” posture); and incoordination and falling when attempting to walk. A smell of ketones may be detectable on the breath. Affected ewes usually become recumbent in 3 to 4 days and remain in a state of profound depression or coma for a further 3 to 4 days, although the clinical course is shorter in fat ewes. Terminally there may be a fetid diarrhea. Fetal death often occurs and is followed by transient recovery of the ewe, but the toxemia caused by the decomposing fetus soon causes a relapse. Affected ewes commonly have difficulty in lambing. Recovery may occur after the ewe lambs or if the lambs are removed by cesarean section in the early stages of the disease. In an affected flock, the disease usually takes the form of a slow, prolonged outbreak, with a few ewes affected each day over a period of several weeks. Recovered ewes may subsequently show a break in the wool.

CLINICAL PATHOLOGY

Hypoglycemia, ketonemia, and ketonuria are characteristic of the disease. The initial changes are similar to ketosis in cattle but the sequel is not. Hypoglycemia can be used as a diagnostic aid in the early stages of the disease, but is of limited value later on when the ewe becomes recumbent, when blood glucose levels may be normal or grossly elevated. This may follow fetal death, which has been shown to remove the suppressing effect of the fetus on hepatic gluconeogenesis. Ketonemia and ketonuria are constant, with serum BHB concentrations greater than 3.0 mmol/L. Sheep develop a severe metabolic acidosis, develop renal failure with a terminal uremia, and become dehydrated. Liver function tests show liver dysfunction. Elevated plasma cortisol concentrations occur, with greater than 10 ng/mL indicative of pregnancy toxemia. However, elevated plasma cortisol can occur with other conditions, such as hypocalcemia.

NECROPSY FINDINGS

Without treatment, pregnancy toxemia in ewes is almost always fatal. At necropsy, there is severe fatty degeneration of the liver and usually constipation, although some

Metabolic Diseases of Ruminants

VetBooks.ir

cases have fetid, light-colored diarrhea. A large single or, more commonly. twin or greater number of fetuses are present. These may have died before the ewe and be in varying stages of decomposition. Histopathologically there is hepatic lipidosis and a poorly defined renal lesion, and there may be evidence of neuronal necrosis. Hepatic glycogen concentrations are usually very low. Concentrations of BHB in the aqueous humor or the CSF greater than 2.5 mmol/L or 5.0 mmol/L respectively, are supportive of a diagnosis of pregnancy toxemia. DIFFERENTIAL DIAGNOSIS Pregnancy toxemia is usually suspected in late-pregnant ewes that show nervous signs and die within 2 to 7 days. There may be a history of exertion, stress, or sudden deprivation of food. Hypocalcemia can occur under similar circumstances, but the following help in differentiation: 1. The onset is within 12 hours of the stress. 2. A considerable proportion of the flock will be affected at the same time. 3. There is obvious myasthenia. 4. It has a much shorter course, 12 to 24 hours. 5. Affected animals respond well to treatment with solutions of calcium salts. Differential diagnoses include • Listeriosis • Cerebral abscess • Acidosis • Uterine torsion or impending abortion • Rabies

TREATMENT Sheep treated very early in the course of the disease generally respond favorably,4 but response to therapy is poor once sheep have become recumbent, and the IV administration of 50% dextrose at this time may hasten death. Optimum therapy requires the cor­ rection of fluid, electrolyte, and acid–base disturbances in addition to treating with glucose. Parenteral Therapy Ideally, individual sheep should be examined biochemically and the corrective therapy based on these results, with fluids, electrolytes, and glucose (dextrose) given over a prolonged period. A recommendation for glucose therapy is the administration of 5 to 7 g of glucose IV 6 to 8 times a day in conjunction with 20 to 40 units of zinc protamine insulin given IM every other day for 3 days. However, in many sheep-raising areas intensive laboratory monitoring and such intensive therapy is not possible because of lack of access, expense, or the number of sheep involved in an outbreak. In the absence of biochemical monitoring, therapy with glucose should be accompanied by the IV injection of isotonic sodium bicarbonate or

lactated Ringer’s solution, with additional fluids given by a stomach tube. Standard doses of corticosteroids have little therapeutic effect in sheep, and thus treatment with these drugs is not recommended, although they are often used. Very large doses are effective in ewes still able to stand, but the success probably rests in the removal of the glucose drain by the induction of premature parturition. Oral Therapy Oral propylene glycol or glycerin (100 mL once daily) can be used to support parenteral glucose therapy. Less intensive therapy with propylene glycol or glycerin alone can give excellent results, especially with early treatment,4 but is less successful with longerstanding cases. Oral drenching every 4 to 8 hours with 160 mL of a commercial calf scours concentrate (containing 28% glucose, 3.9% glycine, 5.3% sodium chloride, and other electrolytes) induces higher blood concentrations of glucose compared with drenching with glycerol or propylene glycol. Reported recovery rates are 90% in early and 55% in advanced cases. For the more intensive treatment of valuable ewes, insulin ([0.4 IU/kg]/d SC), combined with oral glucose precursors and electrolytes, may improve survival compared with treatment with oral glucose precursors and electrolytes alone. Induction of parturition is an option, but it should only be used if the ewe is in the early stage of the disease because there is a delay in the delivery of the lambs (24 hours or more). If the ewe is unlikely to survive this period, cesarean section may be a better option. Induction can be achieved with dexamethasone 21-isonicotinate or the sodium phosphate form, at a dose rate of 16 to 25  mg per ewe, but dexamethasone trimethylacetate appears to be ineffective. Lambs will be born 24 to 72 hours after injection, with most born within 36 hours. Induction of parturition in normal sheep can be achieved with 10  mg of betamethasone or 2.5  mg of flumethasone, but there are no reports of their efficacy in sheep with pregnancy toxemia. Cesarean Section Cesarean section can be used as an alternate to glucose replacement, and provided that ewes are in the early stages of the disease, removal of the lambs by cesarean section probably has the greatest success. The demand for glucose by the lambs is immediately removed, and both the ewe and the lambs have a high chance of survival, provided that the cesarean section is conducted before there is irreversible brain damage in the ewe and the lambs are close to term. If the ewe is recumbent, then chances of survival, for both the ewe and the lamb, are reduced. The lamb may already be dead, and thus ultrasound examination will inform

1725

fetal age and condition and hence whether to undertake a cesarean section.

TREATMENT AND CONTROL Treatment Oral electrolyte and glucose (calf scours) concentrate solution (160 mL qid) (R-1) OR Oral propylene glycol (60 mL bid or 100 mL/d for 3 days) (R-1) For more intensive treatment, include: oral calcium (calcium lactate 12.5 g/d for 3 days); oral potassium (7.5 g KCl/d for 3 days); insulin 0.4 ([IU/kg]/d SC for 3 days) (R-2) If hypoglycemia: Dextrose (60 to 100 mL IV) (R-2) Abort fetus Ewe: Dexamethasone (20 mg IV or IM) (R-2) Doe: Dexamethasone plus prostaglandin F2α (10 mg IM) or synthetic analog (cloprostenol; 75 g/45 kg IM) (R-2) Cesarean section if late-term fetus and valuable ewe/doe (R-2) Control Correct the contributing factors (e.g., insufficient feed or inadequate trough space, intercurrent disease such as foot rot or foot abscess) (R-1)

CONTROL When clinical cases occur, the rest of the flock should be examined daily for evidence of ketosis, and affected animals should be treated immediately with oral glucose/glycine/electrolyte or propylene glycol/glycerol. Supplementary feeding of the flock should immediately be increased or started, with particular attention given to increasing in the intake of energy (carbohydrate). However, care is needed with cereal grains because rapid introduction can cause ruminal acidosis, and ewes may need from 0.25 to 1 kg/ head per day (0.5 to 2.0 lb/head per day). Consequently, good-quality lucerne hay or legume grains, such as lupins or field peas, may be a safer option if ewes are not currently being fed a grain-based supplement, even though ewes do not need the higher protein content of these feeds. Prevention Ensure that the plane of nutrition is rising in the second half of pregnancy, even if it means restricting the diet in the early stages. An ideal condition score for ewes at 90 days of gestation is 2.75 to 3.0 on a scale of 1 to 5. If necessary, ewes with higher condition scores at the end of the first month of pregnancy can be fed to slowly lose 0.5 in condition score during the period to the third month of pregnancy without any detrimental effect on the ewe or the size or viability of the lamb. In many smaller flocks ewes tend to be in

1726

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

excessively high condition score early in pregnancy. The last 2 months are important in the prevention of pregnancy toxemia because 70% of the lamb’s birth weight is gained during the last 6 weeks of pregnancy. In intensively managed flocks the provision of cereal grain or a concentrate containing 10% protein during this period, at the rate of 0.25 kg/d, increasing to 1 kg/d in the last 2 weeks, provides adequate energy. During this period, there should be an increase in body weight of 10% for ewes with single lambs and 18% in those carrying twins, but the average condition score should remain around 3.0. Higher body-condition scores can result in higher birth weight of lambs, but this is usually not a financially viable strategy, and it increases the risk of fat-ewe pregnancy toxemia and dystocia. At the beginning of the fourth month of pregnancy, the flock can be divided into three groups by condition score, suboptimal, acceptable, and excess (overfat), and the groups are then fed accordingly. These can be monitored by condition scoring every 2 to 3 weeks during the fourth and fifth months of pregnancy. Maiden ewes should be fed as a separate group to provide for their growth in addition to pregnancy. Attention should also be given to broken-mouthed or older ewes to ensure that they maintain adequate body condition. There are too many variations in flock structure and husbandry systems to discuss nutritional management in great detail here; readers should consult specialist texts appropriate to the system they work in.5 However, in more intensive systems, especially prime lamb production, ewes can be pregnancy tested by ultrasound and divided into groups depending on whether they are barren or are carrying single or multiple fetuses. Account needs to be taken of those ewes (and does) that are timid and are thus, or for other reasons, slow feeders. If there is insufficient trough space or if the supplement is fed in small amounts and highly edible, a proportion may get little or no feed. The cost-effectiveness of a feeding program should be evaluated. In breeds with low twinning rates that are well managed, it is often more profitable to simply observe the flock and treat the occasional case. Flock monitoring for latent pregnancy toxemia during the last 6 weeks of pregnancy can be conducted using serum BHB; concentrations of 0.8 mmol/L indicate adequate energy intake, 0.8 to 1.6 mmol/L indicate inadequate energy intake, and greater than 1.6 mmol/L indicate severe undernourishment. Pooled samples can reduce the cost of analysis, but serum glucose and BHB concentrations do vary significantly between flocks. Ionophores are used in transition rations for dairy cows to prevent subclinical ketosis. There is some evidence that feeding monensin may have benefits for the energy metabolism of late pregnant ewes. Lower serum BHB,

lowered feed intake, and improved feed efficiency have been observed, and thus further investigation of this strategy is warranted.6 FURTHER READING

Freer M, Dove H, Nolan JV. Nutrient Requirements of Domesticated Ruminants. Collingwood, Australia: CSIRO Publishing; 2007. Radostits O, et al. Pregnancy toxemia in sheep. In: Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1668-1671.

REFERENCES

1. Duehlmeier R, et al. J Anim Physiol Anim Nutr. 2013;97:971. 2. Yarim GF, et al. Vet Res Commun. 2007;31:565. 3. Moallem U, et al. J Anim Sci. 2012;90:318. 4. Cal-Pereyra L, et al. Ir Vet J. 2015;68:25. 5. Freer M, Dove H. Sheep Nutrition. Collingwood, Australia: CABI and CSIRO; 2002. 6. Taghipoor B, et al. Livestock Sci. 2011;135:231.

STEATITIS, PANNICULITIS, AND FAT NECROSIS Steatitis is inflammation of adipose tissue and can affect any fatty tissue. Clinical expression is usually because of inflammation of intraabdominal or subcutaneous fat (panniculitis). The disease can be relatively innocuous or fulminant and is reported for cattle, in which it is referred to as fat necrosis or bovine lipomatosis, and horses. The colloquial name is “yellow-fat disease” because of the color of affected tissues—a result of accumulation of lipofuscin and products of fat oxidation.1-4 The disease is not neoplastic. The disease in cattle is characterized by inflammation and necrosis of fat in the abdominal cavity. It can be clinically silent with lesions detected during rectal examination for pregnancy diagnosis or other reason. Clinical signs of the disease in cattle are usually attributable to space-occupying lesions (such as compression of the rectum) or intestinal obstruction as a result of constriction of the intestine by mesenteric accumulations of fat or fibrotic constriction of the lumen.3 The lesions are firm masses present in any portion of the omental, mesenteric, or retroperitoneal fat or as mobile, free-floating structures in the abdomen.2 The free-floating masses do not appear to originate from necrosis of fat.2 The masses range from small nodules to large, solid, and irregularly shaped tumors. Unlike in horses, in which intraabdominal lipomas are often pedunculated (see Chapter 7) and cause acute intestinal obstruction when the peduncular stalk wraps and constricts the small intestine, the lesions in cattle are seldom pedunculated.2 The clinical disease in cattle can be variable and range from silent through inappetence, decreased milk production, persistent diarrhea, mild recurrent colic, acute colic, dystocia, urinary retention of feces, and decreased passage of feces. Masses can be detected on rectal examination or laparotomy. Ultrasonography (transcutaneous or

transrectal) can be useful in detecting and characterizing the lesions.3 The lesions are present as heterogenous hyperechoic masses in the retroperitoneal, omental, or mesenteric fat. A hyperechoic ring around the kidney is common.3 Affected tissues can be biopsied with ultrasonographic guidance. Abnormalities in the hemogram and serum biochemistry are confined to indi­ cators of inflammation (neutrophilia), hypergammaglobulinemia, decreased concentrations of phospholipids and cholesterol, and an increase in concentration of free fatty acids.3 The disease must be differentiated from lymphosarcoma, adenocarcinoma, intraabdominal abscess, or dry fecal balls in the descending colon. The lesions are composed of necrotic fat embedded in normal adipose tissue with mild inflammatory infiltrates of neutrophils, lymphocytes, plasma cells, macrophages and giant cells, and fibrosis.2 There is rarely evidence of pancreatitis in the disease in cattle.2 The cause of the disease is unknown, although a prevalence of 67% is reported in steers grazing tall fescue, in which serum cholesterol concentrations were abnormally low. The disease in horses affects mostly foals and young animals and ponies. Older animals are less frequently affected.1 Generalized steatitis can be a fulminant disease in horses, ponies, and foals.1,4 Panniculitis, an unusual form of steatitis limited to the subcutaneous tissues, has been reported in an aged pony mare5 and in perivaginal tissues after dystocia.6 Perivaginal steatitis included involvement of the bladder ligament and subsequent rupture of the bladder.6 The clinical signs of generalized steatitis consist of anorexia and depression, fever, tachycardia, and subcutaneous edema.1,4 Painful subcutaneous swellings can occur in the nuchal crest and inguinal and axillary regions. Affected horses often have mild to moderate colic and signs of abdominal tenderness. Rectal examination reveals painful masses in the mesentery of some horses. Hematology and serum biochemical examination reveal mild to moderate leukocytosis, with occasional horses having leucopenia, hypoproteinemia, hypoalbuminemia, and increases in activity in serum of lactate dehydrogenase (LDH), aspartate aminotransferase (AST), GGT, and lipase and amylase.1,4 Serum vitamin E concentrations are sometimes abnormally low. Biopsy of some of the SC swelling reveals histopathological evidence of fat necrosis with mineralization. At necropsy, the fat is hard, dry, and yellow-white, with areas of necrosis forming abscess-like lesions up to 3 cm deep and 10 cm in diameter. The fat lining the abdominal wall may contain firm yellow-white and red tissue nodules up to 3 cm in diameter. Pancreatitis is evident in equids with systemic disease, and there is necrosis and inflammation in most fatty

Metabolic Diseases of Horses

VetBooks.ir

tissues (subcutaneous, retroperitoneal, mesenteric, and omental).1 Generalized steatitis with fat necrosis (“yellow-fat disease”) has been recognized in many species at various ages and is thought to be related to a dietary deficiency of vitamin E and selenium and intake of unsaturated fatty acids.1,5 REFERENCES

1. de Bruijn CM, et al. Equine Vet Educ. 2006;18:38. 2. Herzog K, et al. J Comp Pathol. 2010;143:309. 3. Tharwat M, et al. Can Vet J. 2012;53:41. 4. Waitt LH, et al. J Vet Diagn Invest. 2006;18:405. 5. Radostits O, et al. Steatitis. In: Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2006:1680. 6. Claes E, et al. Vlaams Diergeneeskundig Tijdschrift. 2014;83:36.

Inherited Metabolic Diseases of Ruminants DEFICIENCY OF UMP SYNTHASE (DUMPS) This is a partial deficiency of an enzyme that is involved in the conversion of orotate to uridine 5’-monophosphate (UMP) as a step in the synthesis of pyrimidine nucleotides. It is recorded at a high prevalence in Holstein– Friesian cattle in the United States and Japanese Black cattle and is characterized by an autosomal-recessive form of inheritance and the secretion of high levels of orotate in the milk.1 Heterozygous animals have a partial deficiency of UMP synthase, but they have no individual or herd clinical abnormalities. Heterozygous animals can be detected biochemically by their half-normal levels of erythrocyte UMP synthase or by nested polymerase chain reaction (PCR) testing.2,3 Bovine homozygotes die at about the 40th day of pregnancy. Embryonic mortality is the only form of loss. REFERENCES

1. Ohba Y, et al. J Vet Med Sci. 2007;69:313. 2. Dai Y, et al. China Anim Health Inspection. 2014;31:76. 3. Paiva DS, et al. Genet Mol Res. 2013;12:3186.

HEPATIC LIPODYSTROPHY IN GALLOWAY CALVES Hepatic lipodystrophy has been reported in Galloway calves on five farms in the United Kingdom over a 10-year period. Calves appear normal after birth but die by 5 months of age. Clinically there is tremor, opisthotonus, and dyspnea before affected calves become recumbent and die. At necropsy the liver is enlarged, pale, and mottled. Histologically there is evidence of hepatic encephalopathy. The cause is unknown, but limited evidence suggests a storage disease is possible.

Metabolic Diseases of Horses EQUINE PITUITARY PARS INTERMEDIA DYSFUNCTION (FORMERLY EQUINE CUSHING DISEASE) Equine pituitary pars intermedia dysfunction (PPID) is a slowly progressive neurodegenerative disease of older equids caused by nonmalignant hypertrophy and hyperplasia of melanotropes of the pars intermedia of the pituitary gland. It is characterized in its most severe form by hirsutism, laminitis, polyuria, and polydipsia.

ETIOLOGY

The pars intermedia of equids is composed of a single cell type—melanotropes—which are innervated by dopaminergic neurons of the periventricular nucleus. Innervation by these neurons is inhibitory on secretion by the melanotropes of proopiomelanocortin (POMC)-derived peptides. Thyrotropinreleasing hormone stimulates melanotropes.1 The pars distalis of the pituitary of healthy horses releases adrenocorticotropic hormone (ACTH) in response to, among other stimuli, declines in plasma cortisol concentration. Cortisol exerts negative feedback on secretion of ACTH by the pars distalis, but not by the pars intermedia.2 The disease is attributable to degeneration of the periventricular hypophyseal dopaminergic neurons with subsequent development of a nonmalignant functional tumor comprised of melanotropes of the pars intermedia of the pituitary gland. Cushing syndrome caused by adrenocortical tumors is exceedingly rare in equids.

EPIDEMIOLOGY

The disease is being diagnosed with increasing frequency.3 The prevalence of the disease is not well documented, but surveys of owners indicate hair-coat abnormalities consistent with the disease in 15% to 39% of aged equids. Of 200 randomly selected equids 15 years of age or older in the United Kingdom, 22% had hair-coat abnormalities suggestive of PPID detected on clinical examination,4 and owners of 12% of approximately ~980 aged equids in the United Kingdom reported hair-coat abnormalities and abnormal moulting.5 Similarly, owners of 17% of 974 horses 15 years of age or older in Queensland, Australia, reported hirsutism.6 Given that changes in hair coat are specific (95%) but of unknown sensitivity for the diagnosis of PPID,7 these estimates likely provide the lower range for prevalence of the disease. Accordingly, 21% of 325 randomly selected horses 15 years of age or older in Queensland had PPID diagnosed based on measurement of plasma ACTH concentrations and using

1727

seasonally adjusted cutoff values.8 This likely provides the best current estimate of the prevalence of PPID in mature and aged horses. Reports of prevalence of the disease provided in early studies likely were unreliable as indicators of disease frequency in the overall population of horses because of selective or nonrandom sampling of horses. The disease occurs worldwide in all breeds of horses and ponies. Differences in geographic distribution are not reported. The only well-recognized animal risk factor for the disease is increasing age (adjusted OR of 1.18 [95% CI, 1.1 to 1.25]) per year of age, and there is no apparent sex or breed predisposition.8

PATHOGENESIS

PPID is a neurodegenerative disease in which there is a loss of the inhibitory effect of dopamine with subsequent hypertrophy and hyperplasia of melanotropes of the pars intermedia of the pituitary gland with unchecked secretion of proopiomelanocortin and compression of the neurohypophysis, hypothalamus, and optic chiasma.9 Production of proopiomelanocortin by melanotropes in the pars intermedia is not under the negative feedback control of glucocorticoids, and as a result, affected equids produce large quantities of POMC, melanocytestimulating hormone (α-MSH), β-endorphin, and smaller but still excessive quantities of ACTH. Production of ACTH results in loss of the normal circadian rhythm in serum cortisol concentration.10 The space-occupying effects of the tumor can cause blindness because of compression of the optic chiasm. Polyuria and polydipsia are common and are probably related to neurohypophyseal dysfunction and compression of the pars nervosa, the source of antidiuretic hormone.11 Not all equids with PPID have impaired glucose metabolism.12,13 A proportion of horses, estimated as ~40%, with PPID have evidence of abnormal glucose metabolism, including hyperinsulinemia, hyperglycemia, or both, although only 20% have evidence based on results of an IV glucose and insulin test.12 Furthermore, horses with PPID do not have abnormalities in glucose metabolism detected during an isoglycemic clamp procedure.13 It is unclear if the abnormal glucose metabolism and hyperinsulinemia are attributable to PPID or concurrent equine metabolic syndrome, but it is apparent that there should not be an assumption of abnormalities in glucose metabolism in all equids with PPID.

CLINICAL FINDINGS

Affected equids exhibit one or more findings of hirsutism, hyperhidrosis, polyuria, polydipsia, polyphagia, muscle atrophy (sarcopenia), laminitis, and docile demeanor. Hirsutism is a clinical sign with high specificity (95%) for the disease,7 meaning that aged equids with hirsutism are likely to

1728

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

have the disease and that there will be few false-positive diagnoses when hirsute aged equids are considered to have PPID. Equids with an owner-reported history of hirsutism are 7.8 times (95% CI, 3.7 to 16.6) more likely to have PPID than are nonhirsute equids of similar age.8 Hirsutism is characterized by delayed or absent seasonal moulting resulting in a long, shaggy hair coat. There can be some lightening of the coat color. The changes in hair coat are a result of equids with PPID having a greater proportion of hair follicles in the anagen phase (95% of hair follicles on the neck) than healthy equids (15%).14 Abnormalities of hair follicles resolve and resumption of moulting occurs with administration of pergolide.14 Polyuria and polydipsia are common clinical signs in equids with PPID and are likely secondary to diabetes insipidus and not to hyperglycemia.11 Administration of desmopressin reduces polyuria and polydipsia.11 Hyperhidrosis is reported in affected equids, although it does not appear to have been quantified. Equids with PPID in hot environments can be anhidrotic, and this resolves with treatment of the PPID.15 Myopathy associated with PPID is characterized by atrophy of type 2 (slow-twitch) fiber types consistent with sarcopenia.16 Plasma activity of muscle-derived enzymes is not greater in equids with PPID than in healthy aged-matched equids.16 The molecular basis for muscle atrophy in equids with PPID has been investigated, but the mechanism remains unclear.17 There is often central obesity, characterized by excessive fat deposition in the crest of the neck and in the supraorbital fossae, but this is likely a reflection of comorbidity with equine metabolic syndrome rather than a characteristic of PPID. One report demonstrates insulin resistance in equids with PPID using the euglycemic-hyperinsulinemic clamp technique, but this is not a consistent finding.13,18 However, equids were not screened for hyperinsulinemia before admission to the study and were selected from a population of equids referred for treatment of laminitis, among other diseases. These equids might well have had both equine metabolic syndrome (EMS) and PPID. Further evidence to support this comorbidity is that plasma fructosamine concentrations are not different between nonlaminitic equids with PPID and healthy controls (reference interval of 195.5 to 301.9).19 Equids with PPID and laminitis have plasma fructosamine concentrations that are higher than those of animals with PPID but not laminitis.19 Fructosamine is a reflection of average blood glucose concentrations over a period of weeks, and higher values are indicative of hyperglycemia. Laminitis is common in equids with PPID (see “Laminitis of Horses,” Chapter 15).8 However, it is unclear if this is a result of PPID or comorbidity with EMS.

Rarely, affected equids are blind or have seizures. Affected equids are often infertile and heal poorly. Equids with PPID are considered immunosuppressed and susceptible to development of opportunistic infections and parasitism.20,21 Computed tomography allows measurement of the size of the pituitary gland of equids that correlates well with that measured postmortem.22 The size of the pituitary gland can be evaluated antemortem. The outcome is favorable in that 50% of equids are alive 4.6 years after diagnosis, most owners are satisfied with the equid’s quality of life, and most (28/29; 97%) would treat a second equid with the disease.3 In a study of cases diagnosed between 1993 and 2002, the cause of death among equids (15/20; 85%) was euthanasia, and 11/15 (73%) were euthanized because of conditions associated with PPID.3

CLINICAL PATHOLOGY

There are no characteristic findings on serum biochemical testing or hematology.8 Resting serum cortisol concentrations of affected and healthy equids are similar and not useful in diagnosis.

DIAGNOSTIC CONFIRMATION

Antemortem diagnosis of PPID is not simple and is achieved on the basis of clinical signs and results of one or more of several diagnostic tests. It is important that testing be based on the presence of clinical signs compatible with the disease to minimize the frequency of false-positive diagnoses. Laboratory tests for the disease are not infallible, and the results of these tests should be viewed only in the context of the equid’s clinical signs. Further complicating diagnosis of equine pars intermedia dysfunction is the slow and progressive onset of the disorder. It is therefore likely that attempting a definitive dichotomous answer (disease present or disease absent) based on laboratory testing is unreasonable—some mildly affected equids will test normal, and, less commonly, some apparently healthy equids with histologically normal pituitary glands will test positive. Repeated testing is warranted when test results are ambiguous or not consistent with clinical signs (primarily hirsutism). Assessment of the utility of the various diagnostic tests is prevented by the lack of a gold-standard diagnosis, except for postmortem examination. Determination of sensitivity and specificity of laboratory tests, or clinical signs, is therefore difficult. Furthermore, antemortem testing is complicated by the seasonal and circadian variations in pituitary function with consequent changes in “resting” or basal serum or plasma concentrations of many analytes. Furthermore, plasma concentrations of some analytes, including ACTH, are affected by feeding.23 The changes in pituitary function with season are a recognized physiologic

phenomenon related to preparing or adapting physiologic functions to colder conditions and shorter days.10,24-30 This phenomenon was not generally recognized before about 2005, and reports of the characteristics of diagnostics tests before that date should be interpreted with caution. Laboratory tests used to diagnose pars intermedia dysfunction include measurement of serum or plasma cortisol, ACTH, glucose, or insulin concentrations; the ACTH stimulation test; the thyrotropin-releasing hormone stimulation test; administration of domperidone with subsequent measurement of plasma ACTH; measurement of urinary and salivary corticoid concentrations; and combinations of these tests (Table 17-8). The most widely accepted laboratory tests are the overnight dexamethasone suppression test and measurement of serum ACTH concentration. Other tests have been suggested, but either their sensitivity and specificity have not been determined or they involve measurement of multiple variables or of hormones for which assays are not readily commercially available. Measurement of basal serum insulin concentration is not a useful diagnostic test for equine pars intermedia dysfunction. Measurement of urine or salivary cortisol concentrations has been suggested as a means of diagnosing equine pars intermedia dysfunction, but neither has been validated in a sufficient number of equids to permit assessment of their clinical utility.31 One of the first diagnostic tests developed was the overnight dexamethasone suppression test.31 After collection of a serum sample for measurement of cortisol, dexamethasone (40 µg/kg IM) is administered at about 5 p.m. A second blood sample is collected 15 hours later, with the option to collect a third sample 19 hours after dexamethasone administration. Normal horses will have a serum cortisol concentration of less than 1 µg/dL (28 nmol/L) in the second and third blood samples, whereas affected horses will not show a significant reduction in serum cortisol concentration from that of the initial sample. The sensitivity and specificity of this test are apparently high, with both reported in earlier studies to be approximately 100%.31 However, recent studies of healthy horses demonstrate that there is considerable seasonal variation in the dexamethasone suppression test, with all of 39 healthy aged ponies and horses having normal tests in January (winter) but 10 of the same 39 (26%) having abnormal tests in September (autumn),31 and that the test is specific but not sensitive.32 These results suggest that these diagnostic tests should be interpreted with caution when conducted in the autumn. Measurement of plasma adrenocorticotropin (ACTH) concentration has been widely accepted as a useful laboratory indicator of equine pars intermedia dysfunction. The plasma ACTH concentration varies with the age of the horse and with the season

Metabolic Diseases of Horses

1729

VetBooks.ir

Table 17-8  Diagnostic Testing Methods for Equine pituitary pars intermedia dysfunction (PPID) Diagnostic test

Procedure

Sample

Interpretation

Comments (also see text)

Overnight DEX suppression

Collect serum between 4 and 6 p.m. Administer DEX at 40 µg/kg BW IM. Collect serum 19–20 hours later.

2 serum samples, 1 mL each; 1 pre-DEX administration and 1 post-DEX administration

Serum control of > µg/dL at 19 hours post-DEX administration suggests PPID.

A mildly decreased resting cortisol (pre-DEX administration) is typical of a PPID-affected horse. A resting cortisol of < 1.8 µg/dL is suggestive of iatrogenic adrenal insufficiency.

Endogenous plasma ACTH concentration

Collect EDTA plasma, preferably in plastic blood-collection tube. Separate plasma by centrifugation, and freeze for submission to laboratory. Avoid hemolysis and heat. Process sample within 8 hours of collection.

EDTA plasma sample, 1 mL

Normal reference range depends on methodology and laboratory. Typically an ACTH concentration < 35 pg/mL (chemiluminescent immunoassay) or < 45–50 pg/ mL (radioimmunoassay) is considered normal.

ACTH is likely affected by many biologic events, all of which are not well documented at present. Seasons can have a profound effect, with higher concentrations seen in autumn.

Endogenous plasma α-MSH concentration

Collect EDTA plasma, preferably in plastic blood-collection tube. Separate plasma by centrifugation, and free for submission to laboratory. Avoid hemolysis and heat. Process sample within 8 hours of collection.

1 EDTA plasma sample, 1 mL

Nonautumn reference range: > 35 pmol/L suggests PPID.

Plasma α-MSH concentration is extremely seasonal. High concentrations are observed in autumn.

TRH stimulation assay

Collect serum. Administer TRH, 1 mg IV. Collect serum 30–60 minutes after TRH.

2 serum samples, 1 mL each: pre-TRH administration, 30 minutes post-TRH administration, and 24 hours post-DEX administration

30%–50% increase in serum cortisol 30 minutes after TRH administration suggests PPID.

Pharmaceutical TRH is expensive; TRH compounded for this use may be difficult to obtain. False-positive results may be common.

Combined DEX suppression/ TRH stimulation test

Collect plasma between 8 and 10 a.m. Administer DEX at 40 µg/kg BW IM. Administer TRH, 1 mg IV, 3 hours after DEX administration. Collect serum 30 minutes after TRH and 24 hours after DEX administration.

3 plasma samples, 1 mL each: pre-DEX administration, 30 minutes post-TRH administration, and 24 hours post-DEX administration

Plasma cortisol > 1 µg/dL at 24 hours post-DEX administration or ≥ 66% increase in cortisol levels 3 hours after TRH administration suggests PPID.

Some diagnostic laboratories prefer to use serum for measurement of cortisol levels. The effect of season on the combined test has not been assessed but would likely result in false-positive results as each of the component tests do.

Domperidone response test

Collect EDTA plasma at 8 a.m. Administer domperidone at 3.3 mg/kg BW po. Collect EDTA plasma at 2 and 4 hours after domperidone administration.

3 EDTA plasma samples, 1 mL each

A twofold increase in plasma ACTH concentration suggests PPID.

Higher doses (5 mg/kg po) may improve response. The 2-hour sample is more diagnostic in the summer and autumn, and the 4-hour sample is best in the winter and spring.

Abbreviations: ACTH, adrenocorticotropic hormone; BW, bodyweight; DEX, dexamethasone; IM, intramuscularly; IV, intravenously; THR, thyroid-releasing hormone. (Reproduced, with permission, from McFarlane, D. Vet Clin Equine 2011: 27;93-113. McFarlane D. Vet Clin North Am Equine Pract. 2011;27:93.)

of the year in both the northern and southern hemispheres, but not between ponies and horses.33 The upper reference intervals of plasma ACTH for healthy horses in the United Kingdom in one report were 29 pg/ mL between November and July and 47 pg/ mL between August and October.25 The reference intervals were obtained by sampling a convenience sample of hospitalised horses. A similar pattern is detected in the eastern and southern United States, with the autumnal peak in ACTH occurring earlier in horses in more northern locations.1,24,29,30 This circannual variation in plasma ACTH occurs in

both non-PPID and PPID horses, with PPID horses having higher concentrations than non-PPID horses at all times.10,23,25,26,33,34 Furthermore, the increase in plasma ACTH concentrations stimulated by administration of thyrotropin-releasing hormone (1 mg, IV) to healthy horses is greater in autumn and summer than in late winter.23,27 These results demonstrate the need for including consideration of season (photoperiod) and latitude when assessing the diagnostic importance of plasma ACTH concentrations in aged horses. It is prudent to use reference intervals developed in local laboratories or in distant

laboratories with knowledge of the reference interval for the particular geographic location (latitude) and season of the horse.30 There is no circadian rhythm to ACTH concentrations in horses with PPID, but there is conflicting evidence of a circadian rhythm in healthy horses.10,35,36 It appears that ACTH concentrations of horses are highest at 0800 hours and then decline over the day, although the changes are small and not likely to affect clinical interpretation of plasma ACTH concentrations.35 There is not an ultradial rhythm (periodic changes during a 24-hour period) in plasma ACTH

1730

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

concentration, although measured concentrations do vary over brief periods of time (minutes) and to a greater extent in horses with PPID.35 Fasting and feeding affect plasma ACTH concentrations in healthy horses, with higher concentrations found 2 hours after feeding than after a 12-hour fast (46 vs. 17 pg/mL, respectively).23 Measurement of plasma ACTH combined with use of seasonally adjusted cutoffs provides good sensitivity and specificity for diagnosis of PPID, defined using the presence of hirsutism plus three or more clinical signs as the gold standard.33 The referenced study was of 325 randomly selected horses 15 years of age or older in Queensland, Australia (approximate latitude 27.5°S). Cutoff values for diagnosis of PPID were 30 pg/mL (sensitivity and specificity of 80% and 82%, respectively) for nonautumn months and 77 pg/mL (sensitivity and specificity of 100% and 95%, respectively) during the autumn. It is important to note that the gold standard for determining the characteristics of the test was a clinical examination. Therefore the sensitivity and specificity reported for measurement of plasma ACTH concentration apply only for horses with characteristic clinical signs of the disease. The usefulness of measuring plasma ACTH concentration in horses that have milder, or nonexistent, clinical signs is unknown. Similarly, the clinical importance of elevated ACTH concentrations in younger horses is unclear, and such results should be considered cautiously and carefully before decisions regarding treatment are made. Plasma concentrations of α-melanocytestimulating hormone (α-MSH) correlate well with plasma ACTH concentrations, and comments about seasonal and horse-related factors affecting ACTH concentrations also apply for α-MSH.1,10,24,27,33,37 Plasma ACTH concentrations can be measured before and after administration of thyrotropin-releasing hormone or domperidone.37,38 The thyrotropin-releasing hormone test appears to have greater utility than administration of domperidone, with the latter having greater variation. These tests have not been adequately evaluated to recommend at this time. The combined dexamethasone suppression/thyroid-releasing hormone (TRH) stimulation test has reported sensitivity and specificity of 88% and 76%, respectively.7 The test is performed by administering 40 µg/kg of dexamethasone phosphate (or similar dexamethasone salt) intravenously between 8 a.m. and 10 a.m. Cortisol concentration in serum is then measured 3 hours later, and TRH (1 mg) is administered intravenously. Serum cortisol concentration is measured 30 minutes after TRH administration. Serum cortisol concentrations of healthy horses 30 minutes after TRH administration are unchanged from those at the time of TRH

administration, whereas serum cortisol concentrations in horses with equine pars intermedia dysfunction increase by more than 66% of the baseline value. Plasma fructosamine concentrations do not differ between healthy horses (range 195.5 to 301.9 mu mol/L) and horses with PPID.39 Plasma insulin concentrations are increased in a proportion of horses with PPID and are suggested to be indicative of the risk of laminitis in these horses (see discussion of equine laminitis, Chapter 15).40,41

NECROPSY FINDINGS

The pituitary gland is usually enlarged as a result of the increased numbers of melanocortin cells comprising an adenoma of the pars intermedia. The adrenal cortices are usually of normal width, but they may be thickened in some cases. With the appropriate clinical history, the observation of a welldefined nodule within the pituitary gland is usually sufficient for confirmation of the diagnosis, but histology and immunohistochemical testing of the mass can be performed. There is only fair (kappa = 34%) agreement among pathologists for histologic diagnosis of the disease. DIFFERENTIAL DIAGNOSIS • Insulin resistance • Diabetes insipidus (nephrogenic) • Both of these diseases are exceedingly rare in horses • Obesity • Psychogenic polydipsia or salt eating • Chronic renal failure

TREATMENT Treatment is palliative and not curative in that clinical signs can be controlled by administration of pergolide, but the underlying neurodegenerative disease is not cured. The aim of treatment is to reduce secretion of the products of the melanotropes through the use of dopamine agonists or serotonin antagonists. Treatment must be continued for the life of the horse or pony. The treatment of choice is administration of pergolide mesylate, a dopamine agonist, at 1.7 to 5.5 µg/kg orally every 24 hours. The recommended starting dose is 2.0 to 3.0 µg/kg once daily for 2 months, at which time clinical (hirsutism) and laboratory (plasma ACTH concentration) signs of the disease should be evaluated. The dose can be escalated by 1-µg/kg increments until control of clinical signs is achieved. Pergolide mesylate is rapidly absorbed after oral administration to fasted mares with a time to maximum drug concentration in plasma of 0.4 hours, maximum concentration of 4 ± 2 ng/mL, and terminal elimination half-life estimated to be 5.9 ± 3.4 hours.43 Care should be exercised in the storage of pergolide mesylate compounded in an

aqueous vehicle because it is susceptible to degradation if exposed to heat, light, or both.44 Compounded pergolide formulations in aqueous vehicles should be stored in a dark container, protected from light, and refrigerated, and it should not be used more than 30 days after production. Formulations that have undergone a color change should be considered degraded and discarded.44 A commercial form of pergolide mesylate formulated for use with horses and ponies (Prascend®, Boehringer Ingelheim) is available in some countries. Cyproheptadine, a serotonin antagonist, is administered at 0.25 mg/kg orally every 24 hours for 1 month. If an acceptable response is achieved, then this dose is continued; if not, then the dose is increased to 0.25 mg/kg every 12 hours. This drug is now rarely used in the treatment of PPID. Symptomatic treatment should include clipping of the hair coat in spring, treatment of laminitis and wounds, prevention of injuries and infection, and dietary management to reduce hyperglycemia in those animals with this abnormality documented (see “Equine Metabolic Syndrome”), in addition to maintenance of optimal body weight. Some equids with PPID lose weight and require careful nutritional management.

CONTROL None.

FURTHER READING Durham AE, McGowan CM, Fey K, Tamzali Y, van der Kolk JH. Pituitary pars intermedia dysfunction: diagnosis and treatment. Equine Vet Educ. 2014;26:216-223. McFarlane D. Equine pars intermedia dysfunction. Vet Clin North Am Equine Pract. 2011;27:93-113. McFarlane D. Pathophysiology and clinical features of pituitary pars intermedia dysfunction. Equine Vet Educ. 2014;26:592-598.

REFERENCES

1. McFarlane D, et al. Dom Anim Endocrin. 2006;30:276. 2. McFarlane D. Equine Vet Educ. 2014;26:592. 3. Rohrbach BW, et al. J Vet Int Med. 2012;26:1027. 4. Ireland JL, et al. Equine Vet J. 2012;44:101. 5. Ireland JL, et al. Equine Vet J. 2011;43:37. 6. McGowan TW, et al. Aust Vet J. 2010;88:465. 7. Frank N, et al. J Vet Int Med. 2006;20:987. 8. McGowan TW, et al. Equine Vet J. 2013;45:74. 9. McFarlane D. Ageing Res Rev. 2007;6:54. 10. Cordero M, et al. Dom Anim Endocrin. 2012;43:317. 11. Moses ME, et al. Equine Vet Educ. 2013;25:111. 12. Gehlen H, et al. J Equine Vet Sci. 2014;34:508. 13. Mastro LM, et al. Dom Anim Endocrin. 2015;50:14. 14. Innera M, et al. Vet Dermatol. 2013;24:212. 15. Spelta CW, et al. Aust Vet J. 2012;90:451. 16. Aleman M, et al. Neuromuscul Disord. 2006;16:737. 17. Aleman M, et al. Am J Vet Res. 2010;71:664. 18. Klinkhamer K, et al. Vet Quart. 2011;31:19. 19. Knowles EJ, et al. Equine Vet J. 2013;n/a. 20. McFarlane D, et al. J Vet Int Med. 2008;22:436. 21. McFarlane D, et al. JAVMA. 2010;236:330. 22. Pease AP, et al. J Vet Int Med. 2011;25:1144. 23. Diez de Castro E, et al. Dom Anim Endocrin. 2014;48:77. 24. Beech J, et al. JAVMA. 2009;235:715.

Metabolic Diseases of Horses

VetBooks.ir

25. Copas VEN, et al. Equine Vet J. 2012;44:440. 26. Frank N, et al. J Vet Int Med. 2010;24:1167. 27. Funk RA, et al. J Vet Int Med. 2011;25:579. 28. Haritou SJA, et al. J Neuroendocrin. 2008;20:988. 29. McFarlane D, et al. J Vet Int Med. 2011;25:872. 30. Schreiber CM, et al. JAVMA. 2012;241:241. 31. Radostits O, et al. Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. London: Saunders; 2006:1686. 32. Beech J, et al. JAVMA. 2007;231:417. 33. McGowan TW, et al. Equine Vet J. 2013;45:66. 34. Lee Z-Y, et al. Vet J. 2010;185:58. 35. Rendle DI, et al. Equine Vet J. 2013;n/a. 36. Rendle DI, et al. Equine Vet J. 2014;46:113. 37. Beech J, et al. JAVMA. 2011;238:1305. 38. Beech J, et al. J Vet Int Med. 2011;25:1431. 39. Knowles EJ, et al. Equine Vet J. 2014;46:249. 40. Walsh D, et al. J Equine Vet Sci. 2009;29:87. 41. Durham AE, et al. Equine Vet Educ. 2014;26:216. 42. McFarlane D. Vet Clin North Am Equine Pract. 2011;27:93. 43. Wright A, et al. J Vet Int Med. 2008;22:710. 44. Davis JL, et al. JAVMA. 2009;234:385.

transport into insulin-sensitive cells (adipose tissue, skeletal muscle). EMS is the clinical syndrome, whereas insulin resistance in an underlying metabolic abnormality.

EQUINE METABOLIC SYNDROME

EPIDEMIOLOGY

SYNOPSIS Etiology Unknown, but likely involves genetic predisposition for insulin resistance with phenotypic expression permitted or induced by environmental factors that favor obesity. Epidemiology Associated with obesity and particular breeds, especially ponies. Standardbreds appear to be at reduced risk. No sex predilection. Increasing incidence with age. Clinical signs Obesity, regional adiposity including cresty neck, predisposition to laminitis. Clinical pathology Hyperinsulinemia, hypertriglyceridemia, normal blood glucose concentration. Diagnostic confirmation Demonstration of insulin resistance in presence of clinical signs of equine metabolic syndrome. Measurement of serum insulin and plasma or blood glucose concentrations. Requires validated insulin assay. Treatment Weight loss, which can be difficult to achieve. Exercise. Potentially administration of insulin sensitizing drugs (metformin). Control Maintenance of optimal body condition and prevention of obesity.

Equine metabolic syndrome (EMS) is a recently recognized condition of equids characterized by increased regional adiposity (localized deposition of fat) or generalized obesity, hyperinsulinemia, hypertriglyceridemia, insulin resistance, and a predisposition to laminitis that develops in the absence of other known inciting factors (such as colic, metritis, or acute carbohydrate overload).1 Insulin resistance is defined as abnormally depressed insulin-mediated glucose

ETIOLOGY

EMS likely has important genetic determinants, with manifestation of the syndrome when susceptible animals, by virtue of their genetic composition, are exposed to environmental conditions that favor or enable development of the disease. This is thought to be similar to the situation with human metabolic syndrome or type 2 diabetes. It is speculated that breeds of equids that evolved under conditions of frequent restriction of energy intake, such as during long winters, are genetically predisposed to have efficient energy metabolism that under modern management systems can result in obesity and development of insulin resistance.1,2 The epidemiology of EMS is not well described, and there are few studies that have identified the frequency of the condition, using established case definitions, in large numbers of horses or ponies. Consequently, there is little evidence on outcome (morbidity, case-fatality rates, all-cause morbidity, specific-cause mortality) in horses and only anecdotal information on breed, age, and sex as risk factors. Twenty seven percent (51/188) of ponies of various breeds examined in Australia were hyperinsulinemic (>20 mu/mL) after ponies with documented PPID (see previous section) were excluded.3 There is somewhat more information regarding the epidemiology of obesity in horses, but it should be recognized that not all obese horses are insulin resistant (and therefore do not fit the definition of EMS).4,5 Similarly, there is information on the epidemiology of pasture-associated (endocrinopathic) laminitis (see “Laminitis of Horses,” Chapter 15), and from this one can infer risk factors for insulin resistance and EMS. Horses or ponies of different breeds but similar body weight differ in their insulin resistance, and there is consensus that particular breeds are at increased risk of EMS; these include ponies (of any of the common breeds), Morgan Horses, Paso Fino, Andalusian, Arabian, Saddlebred, Quarter Horses, Tennessee Walking Horses, and Warmblood Horses.1,2 It appears that some light breeds such as Standardbreds and perhaps Thoroughbreds are at reduced risk. The frequency of the condition increases with age in ponies,3 and sex does not appear to be a risk factor. There is a seasonality to the occurrence of pasture-associated laminitis,6 and this might represent seasonal changes in energy intake (from pasture) of susceptible animals rather than seasonal variations in severity of insulin resistance. However, there is evidence that measures of insulin resistance in horses and/or ponies vary depending on season,

1731

with declines in insulin sensitivity of ponies in summer.7 Insulin sensitivity, defined using the combined insulin and glucose intravenous test, and serum insulin concentrations are not related to season in healthy, mature horses.8,9 Obesity is common in domestic horses, with studies in Scotland and the eastern United States finding that 45% and 19%, respectively, of horses were considered obese.10,11 Although interpretation of both studies is limited by the localized nature of the sampling and restrictions on the types of horses included, these studies do support a wider consensus that obesity is common in horses. Chronic intake of energy in excess of maintenance needs (overeating) and insufficient exercise are thought to be risk factors, or inciting factors, for obesity.

PATHOGENESIS

EMS is primarily a manifestation of insulin resistance, and insulin resistance is often, but not always, associated with obesity. The syndrome likely includes abnormalities in energy metabolism, adipocyte function, hemostasis (thrombosis), inflammation, response to lipopolysaccharide (endotoxin) exposure, and oxidant stress.1,12 The pathogenesis of laminitis associated with EMS (“endocrinopathic laminitis”) is discussed elsewhere. Insulin resistance is the decreased rate of transport of glucose into cells of glucosesensitive tissues in response to exposure to insulin. Horses with insulin resistance have lesser reductions in blood glucose concentration in response to administration of insulin than do insulin-sensitive horses.13,14 Insulinstimulated glucose transport is achieved by GLUT-4 (glucose transporter protein 4, which is one of at least 12 glucose transporter proteins) when it is present on the surface of adipose or muscle tissue. Insulin causes the relocation of GLUT-4 within the cell to the cell plasma membrane and subsequent increases in rate of glucose transport into the cell. Horses with insulin resistance have abnormal glycemic and insulinemic responses to oral or IV administration of glucose or oral administration of sugar (see following discussion) and have reduced concentrations of GLUT-4 on the surface of skeletal muscle and adipose tissue.15-17 Insulin resistance in horses is associated with an exaggerated response in plasma/serum insulin concentration after administration of glucose. This exaggerated response allows maintenance of resting blood glucose concentrations in the reference range in affected horses—so-called compensated insulin resistance.1 Insulin resistance in humans is currently thought linked to inflammation induced by macrophage activation in adipose tissue, and there is increasing evidence of a similar mechanism in equids.18 The BCS of horses is also correlated with both plasma insulin concentration and serum amyloid A

1732

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

concentration (an acute-phase protein indicative of inflammation).19 There are only minimal effects of insulin infusion (6-hour duration) on tumor necrosis factor alpha and interleukin(IL)-6 concentrations in the plasma of healthy horses,20 and no association has been found between BCS or insulin concentration and plasma concentrations of tumor necrosis factor and IL-6.19 Horses with EMS have prolonged inflammatory responses (evidenced by plasma IL-8, IL-10 and tumor necrosis factor concentrations) to infusion of endotoxin compared with healthy horses,12 although the clinical importance of this observation is unclear. Horses with EMS have a marked increase in neutrophil reactive oxygen species production induced by phagocytosis that is strongly correlated to the blood insulin concentration.21 In contrast, peripheral blood cells of obese hyperinsulinemic horses showed decreased endogenous proinflammatory cytokine gene expression (IL-1 and IL-6) and similar cytokine response following immune stimulation compared with that of control horses. The authors conclude that this could suggest that, unlike in people, cytokine-mediated inflammation does not increase in direct response to obesity or insulin resistance in horses.21 Mechanisms underlying obesity and regional adiposity are unclear, but at the most fundamental level involve an excess of energy intake over energy expenditure, with deposition of the net excess energy as fat. As discussed previously, some horses and ponies appear to be much more efficient at converting feed into energy, or at regulating energy use, with the result that it is challenging to achieve a reduction in the weight of these animals even with strict control of food intake.22 The genetic or hormonal causes of this resistance to weight loss are unclear, although it is apparent that some horses and ponies have an exaggerated insulinemic response to ingestion of soluble carbohydrate.2,17,23 This exaggerated response could be the underlying mechanism for hyperinsulinemia, obesity or regional adiposity, and endocrinopathic laminitis.24 Adipose tissue is an important source of hormones regulating energy metabolism and of inflammatory cytokines. Obesity, or “overconditioning,” in horses (as assessed by BCS) is associated with higher plasma insulin and leptin concentrations than in optimally conditioned horses.25,26 Obese horses also have higher triglyceride concentrations and lower red blood cell glutathione peroxidase activities (an indication of antioxidant capacity) than optimally conditioned horses.27 Regional obesity is an important risk factor in humans for metabolic syndrome and might be similarly so in horses and ponies. Visceral fat is important in humans, but it appears to be less so in horses, with nuchal ligament fat (which contributes to the cresty neck characteristic of affected horses and ponies) having greater proinflammatory

gene expression (IL-1β and IL-6) in affected horses,28 although others, using measurement of other markers of inflammation, find that the omental and retroperitoneal (visceral) fat of insulin-resistant horses have greater expression of markers of inflammation (Toll-like receptor 4 and suppressor of cytokine signaling 3 [SOCS-3]) than do insulin-sensitive horses.18 Finally, there is evidence of regional differences in glucose transport by adipose tissue, with omental adipose tissue having the highest total GLUT content compared with subcutaneous and nuchal ligament fat in healthy horses, but having a reduced total GLUT4 content and cell surface expression in insulin-resistant horses.16

CLINICAL SIGNS

Equine metabolic syndrome is defined by obesity (with or without regional adiposity), insulin resistance, and increased susceptibility to laminitis. As such, only obesity and regional adiposity and clinical signs of active or past laminitis are physical evidence of the presence of metabolic syndrome. Affected ponies can be hypertensive.5,7 Clinical assessment of obesity/adiposity is achieved by use of body-condition scoring, measurement of subcutaneous or retroperitoneal fat by ultrasound, and grading of regional adiposity. Methods used for research studies include slaughter and dissection with proximate analysis of body constituents or measurement of the deuterium dilution space (volume of distribution) in living animals.29,30 Bioelectrical impedance can be used to measure body-water content, but it has not achieved widespread clinical acceptance.31 Measurement of body weight is not useful for assessment of obesity or adiposity because body weight is highly correlated with height and girth and does not provide an accurate indication of body fat.32 A number of body-condition scoring systems have been developed, and the two most commonly used are those of Henneke (later modified by various authors), which uses a 1-to-9 grading system (Table 17-9 and Fig. 17-7), and Carroll and Huntington, which uses a 1-to-5 grading system.33,34 These grading systems were not developed to assess the fat content (proportion) of horses, but rather to assess “flesh” or the general body condition. Both systems have limitations, including their subjective nature and the fact that they have not been validated in all breeds and body types of horses (validation determines the relationship between BCS and a gold-standard measure of body fat, such as deuterium dilution space or carcass analysis), nor has their reliability (intrarater and interrater agreement/repeatability expressed as an intraclass correlation coefficient or, less optimally, a kappa or weighted-kappa statistic) been demonstrated over large numbers of raters. There are reports of an intraclass correlation coefficient (ICC) of 0.74 for four

raters of 21 mares and 75 ponies, but details are not provided.35 Another reports an ICC of 0.92, but without details to allow assessment of the methodology.25 Additionally, bodycondition scoring systems do not provide an assessment of regional adiposity, which might have greater clinical relevance. BCS (1-to-9 scale) correlates well with percent body fat (TBF) when both are log transformed (eTBF = 0.006 + e1.56*BCS).32 In practical terms, this means that the accuracy of this body-condition scoring system to predict the proportion of body fat declines as BCS increases—for example, the proportion of body fat varies from ~13% to 36% in horses with a BCS of 7 to 8/9.32 The log-log BCS model correctly predicted body fat greater than 20% (BCS = 6.83) in 76% of horses and with sensitivity of 83% and specificity of 71%.32 However, the need to use log-log transforms decreases the general utility of this technique. Body-condition scoring is a demonstrably insensitive measure of changes in body fat—ponies subject to 11% reduction in body weight and a 45% reduction in body fat did not have a change in BCS.36,37 This indicates that the BCS system (1-to-9 scale) has some utility in assessment of body-fat proportion in horses and ponies, but it should be used with a full awareness of its limitations. The BCS correlates well with plasma concentrations of glucose tolerance, insulin sensitivity, and insulin, leptin, and triglyceride concentrations in horses or ponies,5,19,35 all of which could be clinically important. A grading system for assessment of regional adiposity evident as a “cresty neck” has been developed for use with ponies and horses.35 The ICC (a measure of reliability between raters) is 0.70 for four raters of 21 mares and 75 ponies.35 The “cresty neck score” correlates well with plasma insulin and glucose concentrations in pooled horse and pony data, and with insulin, leptin, glucose, and triglyceride concentrations when horses and ponies are considered separately. Additionally, ponies with a cresty neck score of 4 or greater are at increased risk of developing pasture-associated laminitis.39 This scoring system therefore appears to be both reliable (good interrater agreement) and indicative of clinically meaningful variables and outcomes (Table 17-10). Acute laminitis and residual signs of laminitis (sometimes call chronic laminitis) are common in animals with insulin resistance and provide the physical confirmation of EMS in these animals. Clinical signs of laminitis are described under that topic (Chapter 15). Diagnosis Definitive diagnosis of EMS of horses in the field is achieved by demonstration of insulin resistance in equids with appropriate clinical signs (obesity, regional adiposity, laminitis) and is confirmed by measurement of plasma concentrations of glucose and insulin.1

Metabolic Diseases of Horses

VetBooks.ir

Table 17-9  Criteria for estimating body condition in light-breed horses Score

Condition

Description

1

Poor

Animal is extremely emaciated. Spinous processes (parts of vertebrae that project upward), ribs, tailhead, hooks (tuber coxae; hip joints), and pins (tuber ischia; lower pelvic bones) projecting prominently. Bone structure of withers, shoulders, and neck easily noticeable. No fatty tissue can be felt.

2

3

4

Very thin

Thin

Moderately thin

Animal is emaciated. Slight fat covering over base of the spinous processes, transverse processes (portions of vertebrae that project outward) of lumbar (loin area) vertebrae feel rounded. Spinous processes, ribs, tailhead, hooks, and pins are prominent. Withers, shoulders, and neck structures are faintly discernible. Fat is built up about halfway on spinous processes; transverse processes cannot be felt. Slight fat cover over ribs. Spinous processes and ribs are easily discernible. Tailhead is prominent, but individual vertebrae cannot be visually identified. Hook bones appear rounded, but are easily discernible. Fat can be felt around tailhead (prominence depends on conformation). Hook bones are not discernible. Withers, shoulders, and neck are not obviously thin. Negative crease along back (spinous processes of vertebrae protrude slightly above surrounding tissue). Faint outline of ribs is discernible. Fat can be felt around tailhead (prominence depends on conformation). Hook bones are not discernible. Withers, shoulders, and neck are not obviously thin.

5

Moderate

Back is level. Ribs cannot be visually distinguished, but can be easily felt. Fat around tailhead begins to feel spongy. Withers appear rounded over spinous processes. Shoulders and neck blend smoothly into body.

6

Moderately fleshy

May have slight crease down back. Fat over ribs feels spongy. Fat around tailhead feels soft. Fat begins to be deposited along at sides of the withers, behind shoulders, and along neck.

7

Fleshy

May have crease down back. Individual ribs can be felt, but with noticeable filling of fat between ribs. Fat around tailhead is soft. Fat is deposited along withers, behind shoulders, and along neck.

8

Fat

Crease down back. Difficult to feel ribs. Fat around tailhead is very soft. Area along withers is filled with fat. Area behind shoulder is filled with fat and flush with rest of the body. Noticeable thickening of neck. Fat is deposited along inner thighs.

9

Extremely fat

Obvious crease down back. Patchy fat appears over ribs. Bulging fat around tailhead, along withers, behind shoulders, and along neck. Fat along inner thighs may rub together. Flank is filled with fat and flush with rest of the body.

Based on Henneke et al. (1983) Henneke DR, et al. Equine Vet J. 1983;15:371 and reproduced with permission. Carter RA, et al. In: Geor RJ, et al., eds. Equine Applied and Clinical Nutrition. W.B. Saunders; 2013:393.

Insulin resistance can be detected by use of measurement of insulin and glucose concentrations in a single blood sample (point testing) or by more sophisticated testing using measurement of these analytes on multiple occasions after administration of glucose and insulin to equids (dynamic testing)—either as the euglycemic clamp technique or the frequently sampled intravenous glucose and insulin test (minimal model).40-42 The former has greater utility in clinical and field settings, although with the potential for reduced sensitivity and/or specificity, whereas the latter is useful for research or referral settings and is regarded as the gold standard for diagnosis. Hyperglycemia is rarely detected in equids with insulin resistance, and

measurement of blood glucose concentrations alone is not a useful test for detection of insulin resistance.1 Detection of persistent, inappropriate hyperglycemia (i.e., that not associated with stress or ingestion of food) should prompt consideration of diabetes mellitus.43 Hyperinsulinemia in the absence of conditions that increase insulin secretion (stress, pain, feeding) is strong evidence of the presence of insulin resistance.1 Interpretation of plasma or serum insulin concentration must include consideration of a number of factors that could affect the actual concentration reported by the laboratory. Physiologic factors that can increase serum insulin concentration include feeding, stress and pain, and administration of alpha-2

1733

Table 17-10  Grading system for assessing regional adiposity in the neck of ponies and horses Score

Description

0

No visual appearance of a crest (tissue apparent above the ligamentum nuchae). No palpable crest.

1

No visual appearance of a crest, but slight filling felt with palpation.

2

Noticeable appearance of a crest, but fat deposited fairly evenly from poll to withers. Crest easily cupped in one hand and bent from side to side.

3

Crest enlarged and thickened, so fat is deposited more heavily in middle of the neck than toward poll and withers, giving a mounded appearance. Crest fills cupped hand and begins losing side-to-side flexibility.

4

Crest grossly enlarged and thickened and can no longer be cupped in one hand or easily bent from side to side. Crest may have wrinkles/creases perpendicular to topline.

5

Crest is so large it permanently droops to one side.

adrenergic agonists (xylazine, detomidine, romifidine, etc.). Feeding increases plasma insulin concentration in both healthy and insulin-resistant horses and confounds interpretation of the results of testing. Pain and stress increase both plasma glucose and insulin concentrations through increases in cortisol and epinephrine concentrations in blood, which decrease insulin sensitivity.44 This could be important when testing equids with active laminitis or other causes of pain—evaluation should be delayed until the pain is resolved.1 Point testing of plasma glucose and insulin concentrations should be performed under controlled conditions and ideally after 6 hours of feed withholding and preferably with sampling between 8 and 10 a.m.1 Horses can be fed a small amount of hay with a low content of nonstructural carbohydrates the night before testing (approximately 2 kg of hay per 500-kg horse) and then nothing immediately before testing.1 Laboratory factors can influence the insulin concentration reported for a blood sample. This is primarily a result of differences in testing methodology returning different concentrations for the same blood sample.45 Until recently, most testing for equine insulin involved use of kits or testing methodology designed for use with human samples, taking advantage of the

1734

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Fig. 17-7  Appearance of light-breed horses at each of the body-condition scores described by Henneke et al. (1983). Henneke DR, et al. Equine Vet J. 1983;15:371. (Reproduced with permission from Carter RA, et al. In: Geor RJ, et al., eds. Equine Applied and Clinical Nutrition. W.B. Saunders; 2013:393.)

considerable cross-reactivity between human and equine insulin. Equine-specific tests are now available, and their precision, accuracy, and specificity have been reported.45 The gold-standard methodology is liquid chromatography–mass spectrometry (LC-MX), but this is expensive and has limited accessibility in laboratories processing large numbers of clinical samples. Analysis of six commonly used or available test kits for measuring plasma or serum concentrations of insulin demonstrated that none reflected concentrations measured by LC-MS and that only one provided reliable (valid) results— the Siemens Coat-a-Count Insulin Radioimmunoassay (RIA)—and only if samples with concentrations exceeding the highest standard were diluted with charcoal-stripped plasma and not the provided diluents.45

The effect of use of differing methods of measuring serum insulin concentration is that use of “cutoff ” values for detecting insulin resistance provided by different laboratories is problematic. A value of 20 µU/mL is recommended as a cutoff, measured using the Siemens Coat-a-Count Insulin RIA, for defining insulin resistance.1 However, this value should be interpreted with caution because the sensitivity (proportion of false negatives) and specificity (proportion of false positives) are not reported, and the test as a way of defining insulin resistance has not been well validated. It can be a useful screening test.1 Local laboratories should be contacted before testing to determine the test used and whether the laboratory has determined reference ranges for its testing methodology.

Proxy indicators of insulin resistance, derived from measurement of plasma glucose and insulin concentrations, have been proposed and used to define insulin resistance and predict predisposition to laminitis in ponies.7,40,41,46 A commonly used proxy is the RISQI: RISQI = insulin concentration−0.5 where lower values of the RISQI indicate lower insulin sensitivity. Dynamic testing is usually achieved using combined glucose and insulin challenge tests of varying complexity.1,41 One of these tests measures the insulin and glucose responses to IVs administration of glucose (150 mg/kg BW) and insulin (0.10 U/kg) with frequent sampling of blood (immediately before and at 1, 5, 15, 25, 35, 45, 60, 75,

Metabolic Diseases of Horses

VetBooks.ir

90, 105, 120, 135, and 150 minutes after infusion).1,5 Blood glucose concentrations of insulin-sensitive horses return to baseline values within 45 minutes of infusion of glucose. Insulin-resistant horses have a delayed decline of blood glucose concentrations and an exaggerated increase in plasma/ serum insulin concentrations.1,5 A more complex test, involving the frequent sampling of blood and delayed administration of insulin, is analyzed using the “minimal model” and yields four measures of insulin sensitivity, glucose disposal, and insulin secretion (pancreatic response).41 Dynamic testing can also involve the administration of insulin and monitoring of blood (plasma) glucose concentrations.13,14 More complex testing involves the IV administration of increasing doses of insulin (either bovine or human recombinant) and measurement of blood glucose concentrations at defined times. The dose of insulin required to achieve a 50% reduction in blood glucose concentration is then used as the diagnostic index. This test is cumbersome and has a high risk of inducing hypoglycemia and is therefore of limited clinical utility.13 A modified test involves administration of 0.1 U/kg IV of recombinant human insulin and measurement of blood glucose concentrations immediately before and 30 minutes after.14 Insulin-sensitive horses (n = 6 in the study) all had a 50% or greater reduction in blood glucose concentrations 30 minutes after insulin administration, whereas none of the insulin-resistant horses (n = 6) had this response. The sensitivity and specificity of this test need to be determined in larger numbers of healthy and insulin-resistant horses for it to be clinically useful. An oral sugar test (OST) provides results as reliable as the IV glucose tolerance test for detection of insulin resistance and hyperinsulinemia. The test involves oral administration of 0.15 mL/kg of corn syrup (approximately 150 mg/kg BW of dextrose-derived digestible sugars) to equids after an overnight fast. Plasma insulin and glucose concentrations are measured immediately before and 30, 60, and 90 minutes after administration of corn syrup. Equids with EMS have higher glucose and insulin concentrations than do unaffected horses.17

CLINICAL PATHOLOGY

Clinical pathology includes detection of hyperinsulinemia (as noted in the previous discussion), hyperleptinemia, hypertriglyceridemia, and normoglycemia. Plasma ACTH concentrations of horses with EMS are within the reference range for healthy horses, noting that horses with EMS might also have PPID.

NECROPSY

Affected horses are usually examined postmortem because of laminitis. Other than signs of laminitis and obesity/regional

adiposity, there are no other characteristic lesions. The pancreas is normal. DIFFERENTIAL DIAGNOSIS • Obesity without insulin resistance • Pituitary pars intermedia dysfunction • Laminitis of cause other than EMS (systemic septic disease such as colitis or metritis)

TREATMENT The objective of treatment is to improve the insulin sensitivity of affected equids. This can be achieved by reducing the proportion of the body made up of fat through careful dietary control. Administration of insulinsensitizing drugs is attracting interest, but has yet to be of clearly demonstrated clinical utility. Dietary Management The fundamental aims of dietary management are as follows: • Achieve and maintain an ideal BCS (body weight, noting earlier comments that body weight does not indicate body composition). • Minimize intake of nonstructural carbohydrates because these induce an insulinemic and hyperglycemic response in EMS-affected equids and at-risk animals.1 • Ensure an adequate and balanced intake of essential nutrients. The ideal BCS of ponies has been much mentioned, but little or seldom defined. The ideal body condition for a pony or horse would be one at which it has insulin sensitivity within the reference range (i.e., is not insulin resistant), has plasma triglyceride concentrations within the reference range, and is not at increased risk of laminitis. There is likely no one BCS that meets all of these criteria for all equids, and each animal must be considered in light of its own circumstances and physiology. Monitoring of measures of insulin resistance and plasma triglyceride concentration would provide guidance in achieving the animal’s ideal, or acceptable, body weight. Achieving and maintaining an acceptable BCS (as a proxy for proportion of body fat) is not easy.47 Animals at most risk of obesity and regional adiposity are often metabolically equipped to maintain this body condition, and reducing body weight or BCS might not be as simple as just reducing feed intake.22 Recommendations for reduction in feed intake suggest that feeding 1.25% of body weight daily as hay is adequate to achieve a gradual reduction in body weight. However, planning a diet to provide a reduction in body weight while ensuring an adequate intake of essential nutrients and providing for the digestive and psychological health of the horse or pony can be challenging.

1735

An initial step must be to eliminate or severely reduce access to pasture. Pasture provides unregulated access to fodder, and ponies can consume 2% to 5% of their body weight in pasture each day. This will result in weight gain. Additionally, pasture, and especially green pasture, has a high content of nonstructural carbohydrates (glucose, starch) that induces a glycemic and hyperinsulinemia response in ponies and susceptible horses. Access to pasture can be eliminated by housing animals on a dry lot or by fitting a grazing muzzle. Hay can be fed as 1.25% to 1.5% of body mass as dry matter per day. The nonstructural carbohydrate content of the hay should be reduced by soaking it in cold water for 12 to 16 hours before feeding. The water used to soak the hay should not be provided to the horse or pony. Feeding this diet will induce a reduction in body weight and improvement in indices of insulin sensitivity.22,37,48,49 Individual horses and ponies can be resistant to weight loss, and a reduction in daily dry matter intake to 1% of body weight might be needed to achieve loss of body weight. However, restriction of intake to this level can cause behavioral changes, such as eating bedding, chewing tails of companions, and other allotriophagia. An early example of dietary restriction inducing allotriophagia was that of Robert Falcon Scott’s use of ponies in an expedition to reach the South Pole. The ponies displayed profound appetite for roughage as a consequence of their highly concentrated ration. Supplements including chromium have been promoted as improving insulin sensitivity in horses. There is evidence that they are not effective, and there is no evidence of efficacy.50 Exercise Exercise increases the insulin sensitivity of horses,51 and it appears sensible to recommend an increase in the amount of exercise of obese ponies and horses.1 However, moderate exercise training does not improve the insulin sensitivity of horses.52 Moderate exercise by nonobese ponies previously affected with laminitis reduced serum amyloid A concentrations, heptoglobin concentrations, and postexercise serum insulin concentrations.53 These results suggest a beneficial role for relatively lowintensity exercise (10 minutes enforced walking followed by 5 minutes of trotting) in reducing inflammation in ponies at risk of laminitis. Medications Administration of levothyroxine or metformin has been advocated for treatment of EMS.1,54,55 Levothyroxine (0.1 mg/kg orally q24 h) causes weight loss and improves insulin sensitivity in obese horses and is recommended as an adjunct to dietary management.55

1736

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Metformin, which is used for treatment of type 2 diabetes in people, has been administered to horses in an attempt to improve insulin sensitivity. Although initial reports were favorable,56 more recent pharmacologic investigation has not demonstrated its efficacy at a dosage of 15 mg/kg orally q12h in improving insulin sensitivity.54,57-59 However, administration of metformin at 30 mg/kg q12h reduced glycemic and insulinemic responses of healthy horses and horses with dexamethasone-induced insulin resistance to administration of dextrose.60 Whether this dosage will be effective in horses or ponies with naturally occurring insulin resistance remains to be determined. Pioglitazone has been investigated in healthy horses, in which it does not improve insulin sensitivity (at 1 mg/kg q24h for 12 days) or attenuate the effects of endotoxin infusion on indicators of systemic inflammation.61,62 The pharmacokinetics of pioglitazone in horses have been reported.63 FURTHER READING

Frank N, Tadros EM. Insulin dysregulation. Equine Vet J. 2014;46:103-112. Frank N, et al. Equine metabolic syndrome. J Vet Int Med. 2010;24:467-475.

REFERENCES

1. Frank N, et al. J Vet Int Med. 2010;24:467. 2. Bamford NJ, et al. Dom Anim Endocrin. 2014;47: 101. 3. Morgan RA, et al. Aust Vet J. 2014;92:101. 4. Treiber KH, et al. JAVMA. 2006;228:1538. 5. Frank N, et al. JAVMA. 2006;228:1383. 6. Menzies-Gow NJ, et al. Vet Rec. 2010;167:690. 7. Bailey SR, et al. Am J Vet Res. 2008;69:122. 8. Funk RA, et al. J Vet Int Med. 2012;26:1035. 9. Place NJ, et al. J Vet Int Med. 2010;24:650. 10. Thatcher CD, et al. J Vet Int Med. 2012;26:1413. 11. Wyse CA, et al. Vet Rec. 2008;162:590. 12. Tadros EM, et al. Am J Vet Res. 2013;74:1010. 13. Caltabilota TJ, et al. J Anim Sci. 2010;88:2940. 14. Bertin FR, et al. Dom Anim Endocrin. 2013;44:19. 15. Waller AP, et al. J Vet Int Med. 2011;25:315. 16. Waller AP, et al. Biochim Biophys Acta. 2011;1812:1098. 17. Schuver A, et al. J Equine Vet Sci. 2014;34:465. 18. Waller AP, et al. Vet Immunol Immunopathol. 2012;149:208. 19. Suagee JK, et al. J Vet Int Med. 2013;27:157. 20. Suagee JK, et al. Vet Immunol Immunopathol. 2011;142:141. 21. Holbrook TC, et al. Vet Immunol Immunopathol. 2012;145:283. 22. Argo CM, et al. Vet J. 2012;194:179. 23. Borer KE, et al. J Anim Sci. 2012;90:3003. 24. Bailey SR, et al. Anim Prod Sci. 2013;53:1182. 25. Carter RA, et al. Am J Vet Res. 2009;70:1250. 26. Ungru J, et al. Vet Rec. 2012;171:528. 27. Pleasant RS, et al. J Vet Int Med. 2013;27:576. 28. Burns TA, et al. J Vet Int Med. 2010;24:932. 29. Dugdale AHA, et al. Equine Vet J. 2011;43:552. 30. Dugdale AHA, et al. Equine Vet J. 2011;43:562. 31. Latman NS, et al. Res Vet Sci. 2011;90:516. 32. Dugdale AHA, et al. Vet J. 2012;194:173. 33. Carroll CL, et al. Equine Vet J. 1988;20:41. 34. Henneke DR, et al. Equine Vet J. 1983;15:371. 35. Carter RA, et al. Vet J. 2009;179:204. 36. Dugdale AHA. Equine Vet J. 2011;43:121. 37. Dugdale AHA, et al. Equine Vet J. 2010;42:600.

38. Carter RA, et al. In: Geor RJ, et al., eds. Equine Applied and Clinical Nutrition. W.B. Saunders; 2013:393. 39. Carter RA, et al. Equine Vet J. 2009;41:171. 40. Kronfeld D. J Equine Vet Sci. 2006;26:281. 41. Kronfeld DS, et al. JAVMA. 2005;226:712. 42. Firshman AM, et al. Equine Vet J. 2007;39:567. 43. Durham AE, et al. Equine Vet J. 2009;41:924. 44. Tiley HA, et al. Am J Vet Res. 2007;68:753. 45. Tinworth KD, et al. Dom Anim Endocrin. 2011;41:81. 46. Borer KE, et al. Equine Vet J. 2012;44:444. 47. Frank N, et al. Equine Vet J. 2014;46:103. 48. McGowan CM, et al. Vet J. 2013;196:153. 49. Schmengler U, et al. Livestock Sci. 2013;155:301. 50. Chameroy KA, et al. Equine Vet J. 2011;43:494. 51. Stewart-Hunt L, et al. Equine Vet J. 2010;42:355. 52. Carter RA, et al. Am J Vet Res. 2010;71:314. 53. Menzies-Gow NJ, et al. Equine Vet J. 2013;n/a. 54. Durham AE. Vet J. 2012;191:17. 55. Frank N, et al. Am J Vet Res. 2005;66:1032. 56. Durham AE, et al. Equine Vet J. 2008;40:493. 57. Tinworth KD, et al. Vet J. 2012;191:79. 58. Tinworth KD, et al. Vet J. 2010;186:282. 59. Tinworth KD, et al. Am J Vet Res. 2010;71:1201. 60. Rendle DI, et al. Equine Vet J. 2013;45:751. 61. Wearn JG, et al. Vet Immunol Immunopathol. 2012;145:42. 62. Suagee JK, et al. J Vet Int Med. 2011;25:356. 63. Wearn JMG, et al. J Vet Pharmacol Ther. 2011;34:252.

PHEOCHROMOCYTOMA (PARAGANGLIOMA) Pheochromocytomas are unusual tumors of domestic animals and occur in cattle, sheep, and horses.1-5 A pheochromocytomas is a neuroendocrine tumor of chromaffin cells of the adrenal medulla or extraadrenal chromaffin tissue. The tumor secretes catecholamines; in humans, clinical signs are related to elevated concentrations of circulating epinephrine or norepinephrine. The clinical presentation in horses usually involves intermittent or acute colic or hemoabdomen. Horses can die acutely of exsanguination into the abdomen from ruptured tumor.2 Affected horses can be persistently or intermittently tachycardic with excessive or untimely sweating. The mass can be palpable near the left kidney or imaged by transrectal ultrasonography.2,6 The normal right adrenal gland cannot be imaged transrectally in a horse.6 The disease in cattle and sheep is usually detected at postmortem examination and has no real economic impact, with the exception of the rare valuable bull affected.5 The disease can occur as part of multiple endocrine neoplasia, but is usually solitary, although gangliomas can metastasise.7 Antemortem diagnosis can be confirmed by measuring high concentrations of metanephrine and vanillylmandelic acid, both of which are metabolites of catecholamines, in blood or urine. There is no effective treatment, nor are there control measures. REFERENCES

1. Aydogan A, et al. Rev Med Vet. 2012;163:536. 2. Elsar N, et al. Israel J Vet Med. 2007;62:53.

3. Germann SE, et al. Vet Rec. 2006;159:530. 4. Nielsen AB, et al. J Comp Pathol. 2012;146:58. 5. Seimiya YM, et al. J Vet Med Sci. 2009;71:225. 6. Durie I, et al. Vet Radiol Ultrasound. 2010;51:540. 7. Herbach N, et al. J Comp Pathol. 2010;143:199.

GLYCOGEN BRANCHING ENZYME DEFICIENCY IN HORSES Glycogen branching enzyme deficiency (GBED) is a fatal condition of fetuses and neonatal foals of the Quarter Horse, Paint Horse, and associated breeds.1 The disease is caused by a nonsense mutation in codon 34 of the GBE1 gene, which prevents the synthesis of a functional GBE protein and severely disrupts glycogen metabolism.1 The mutant GBE1 allele frequency in registered Quarter Horse, Paint Horse, and Thoroughbred horses is reported as 0.041, 0.036, and 0.000, respectively.2 Among 651 eliteperformance American Quarter Horses, 200 control American Quarter Horses, and 180 control American Paint Horses, the GBED allele was detected with an overall frequency of 0.054.3 GBED is inherited as a simple recessive trait from a single founder.2 The disease is reported in North America and Germany.4 Affected foals are aborted, born dead, or affected at birth. It is estimated that up to 2.5% of fetal and early neonatal deaths in Quarter Horses and related breeds are associated with this defect.2 Foals that are born alive are weak and hypothermic, some have flexural limb deformities, and all die usually within hours to days of birth.5 Affected foals have refractory hypoglycaemia and minor elevations in serum activity of creatine kinase. The disease is confirmed by detection of periodic acid–Schiff (PAS)-positive inclusions in the cardiac or skeletal muscle and genotype analysis. There is no effective treatment, and control is by selective and prudent breeding. REFERENCES

1. Ward TL, et al. Mamm Genome. 2004;15:570. 2. Wagner ML, et al. J Vet Int Med. 2006;20:1207. 3. Tryon RC, et al. JAVMA. 2009;234:120. 4. Winter J, et al. Pferdeheilkunde. 2013;29:165. 5. Finno CJ, et al. Vet J. 2009;179:336.

LACTATION TETANY OF MARES (ECLAMPSIA, TRANSPORT TETANY) Lactation tetany of mares is caused by hypocalcemia and is characterized by abnormal behavior progressing to incoordination and tetany. The precise cause of the hypocalcemia has not been determined, but the cause of the clinical signs is a marked reduction in serum concentration of ionized calcium. The effect of feeding diets high in calcium, such as alfalfa hay, during late pregnancy, and of abrupt changes in diet after parturition, have not been investigated in horses

Metabolic Diseases of Horses

VetBooks.ir

as they have in cattle (see discussion of milk fever). The disease was most common when draft-horse breeding was widely practiced, but it is uncommon now.1 The case fatality rate is high in untreated animals. Most cases occur in lactating mares, either at about the 10th day after foaling or 1 to 2 days after weaning. High-producing mares grazing on lush pasture are most susceptible and in many instances are engaged in hard physical work. The housing of wild ponies or prolonged transport can precipitate an episode. The latter has been a particularly important factor in the etiology of the disease in Britain and has been credited with precipitating it even in stallions and dry mares. Occasional cases occur without there being any apparent cause. The disease has occurred in a 20-yearold gelding pony. Hypocalcemia with clinical signs also occurs in horses used for prolonged exercise, such as endurance racing or 3-day events. Hypocalcemia occurs in other diseases of horses, including colic and colitis, and as a result of hypoparathryoidism.2,3 Many mild cases of lactation tetany that recover spontaneously occur after transport, but the case fatality rate in some shipments can be greater than 60%. Mares that develop the disease at the foal heat or at weaning are usually more seriously affected, and the case fatality rate is high if mares are not treated in a timely fashion. Severely affected animals sweat profusely and have difficulty moving because of tetany of the limbs and incoordination. The gait is stiff, and the tail is slightly raised. Rapid, labored respirations and wide dilatation of the nostrils are often accompanied by synchronous diaphragmatic flutter (“thumps”) evident as a distinct thumping sound from the thorax. Muscular fibrillation, particularly of the masseter and shoulder region, and trismus are evident, but there is no prolapse of the membrana nictitans. Affected animals are not hypersensitive to sound, but handling can precipitate increased tetany. The temperature is normal or slightly elevated, and although the pulse is normal in the early stages, it later becomes rapid and irregular. The mare might make many attempts to eat and drink but appears to be unable to swallow, and passage of a stomach tube can be difficult. Urination and defecation are in abeyance, and peristalsis is reduced. Within about 24 hours the untreated mare becomes recumbent; tetanic convulsions develop and become more or less continuous. The mare dies about 48 hours after the onset of illness. The tetany and excitement in the early stages suggest tetanus, but there is no prolapse of the third eyelid, and there is the usual relationship to recent foaling or weaning and physical exertion. The anxiety and muscle tremor of laminitis can be confused with those of lactation tetany, especially when it

occurs in mares that have foaled and retained the placenta. Pain in the feet and bounding digital pulses are diagnostic features of this latter disease. Hypocalcemia occurs with serum concentrations in the range of 4 to 6 mg/dL (1 to 1.50 mmol/L), and the degree of hypocalcemia has been related to the clinical signs. When serum calcium levels are higher than 8 mg/dL (2 mmol/L), the only sign is increased excitability. At levels of 5 to 8 mg/ dL (1.25 to 2 mmol/L), there are tetanic spasms and slight incoordination. At levels of less than 5 mg/dL (1.25 mmol/L), there is recumbency and stupor. It is the con­ centration of ionized calcium that is important, and some animals, such as horses used for 3-day events, can have normal total calcium concentrations but abnormally low ionized calcium concentrations as a result of changes in acid : base status. If possible, serum concentrations of ionized calcium should be measured in horses with clinical signs suggestive of hypocalcemia. Hypomagnesemia with serum magnesium levels of 0.9 mg/dL (0.37 mmol/L) has been observed in some cases, but only in association with recent transport. Hypermagnesemia has been reported in other cases. Treatment by IV administration of calcium borogluconate as recommended in the treatment of parturient paresis in cattle results in rapid, complete recovery. The dose for a 500-kg mare is 300 to 500 mL of a 25% solution of calcium borogluconate or gluconate administered slowly (over 15 to 30 min) intravenously. One of the earliest signs of recovery is the voiding of a large volume of urine. Occasional cases that persist for some days have been recorded. REFERENCES 1. Radostits O, et al. Lactation tetany of mares. In: Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2006:1651. 2. Borer KE, et al. Equine Vet Educ. 2006;18:320. 3. Durie I, et al. J Vet Int Med. 2010;24:439.

EQUINE HYPERLIPEMIA SYNOPSIS Etiology Abnormal energy metabolism secondary to inadequate caloric intake. Epidemiology Pregnant or lactating middle-aged, overweight ponies, donkeys, and American miniature horses. Worldwide. Sporadic. Clinical signs Depression, anorexia, weight loss, ventral edema, muscle fasciculation, mania, recumbency. Clinical pathology Hypertriglyceridemia (triglyceride > 500 mg/dL, 5 mmol/L). Necropsy findings Widespread lipidosis, swollen liver, hepatic rupture.

1737

Treatment Increase energy intake through enteral or parenteral feeding. Treat underlying disease. Control Maintain optimal body condition. Prevent disease and nutritional stress, including changes in diet and prolonged transportation.

ETIOLOGY The potentially life-threatening disease hyperlipemia is associated with hyperlipidemia (an abnormal concentration of lipids in blood) in equids. The disease is a result of a derangement in fat metabolism secondary to nutritional stress and, in particular, inadequate energy intake.1 Hyperlipemia is the clinical syndrome of depression, weakness, and ventral edema with high blood concentrations of triglycerides and hepatic lipidosis. It carries a high case-fatality rate. A related condition is the detection of hypertriglyceridemia associated with an overt, severe, primary disease (colic, neoplasia, endocrine disease) in which the triglyceridemia likely has minimal clinical importance. Hyperlipemia has its greatest importance as a disease of ponies and donkeys in field situations and related to relatively minor inciting causes.

EPIDEMIOLOGY Occurrence Hyperlipemia occurs worldwide. Although its occurrence is sporadic, multiple cases can occur on a farm when there are a number of at-risk animals exposed to the same inciting factor, such as lack of adequate grazing or supplementary feeding. The annual incidence of the disease in ponies in southeastern Australia is 5%, and it is 2% to 10% in donkeys in the United Kingdom.2 The case-fatality rate is 40% to 80%, although it appears to be less in hospitalized equids provided more focused care.2,3 Incidence varies with season and locality; the disease in ponies in Europe occurs most commonly during late gestation (January–March), whereas in southern Australia, the disease is more common in ponies during early lactation (November–January). Animal Risk Factors Hyperlipemia can occur in any breed of horse or pony and in donkeys, but it is more common in ponies and donkeys.2,3 Any breed of equid can develop hypertriglyceridemia as a result of a primary disease, but development to the clinical condition lipemia is most common in ponies, miniature horses, miniature donkeys, and donkeys. The disease is considered most common in females, uncommon in pony stallions and geldings, and rare in foals. Most affected ponies are more than 4 years old, and the peak incidence occurs in 9-year-olds. Hypertriglyceridemia occurs in foals secondary to other diseases.3-5

1738

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

Risk factors for hyperlipemia (triglyceride concentration ≥ 4.4 mmol/L) in 449 donkeys with the disease from a population of 3829 donkeys included concurrent disease (odds ratio [OR] 77, 95% confidence interval [CI], 45 to 129), weight loss in previous month (OR = 6.4, 3.6 to 11.3), relocation to a new site (farm) (OR = 3.9, 1.3 to 12), dental disease (OR = 1.7, 1.1 to 2.8), history of inappetence (OR = 3.2, 1.3 to 7.9), and increasing age (OR = 1.26, 1.1 to 1.45).2 Horses and ponies with primary endocrine disease, including pituitary pars intermedia dysfunction (PPID) or suspected diabetes mellitus and conditions associated with insulin resistance, can have marked elevations in serum triglyceride concentrations (10.5 to 60.3 mmol/l) without evident clinical signs attributable to hypertriglyceridemia. The hypertriglyceridemia resolves with successful treatment of the underlying endocrine disease.6 Pregnancy and lactation increase the risk of the disease in ponies, but not in donkeys. The disease in miniature horses is always associated with underlying disease, such as colic, which is apparently an important risk factor. Underlying disease is identified in 50% of cases in ponies and 72% of affected donkeys;2 however, many cases occur in pregnant or lactating pony mares without evidence of other disease. Overweight ponies and donkeys are at increased risk, and insulin resistance is likely a risk factor for the disease.7 Onset of disease is often preceded by some sort of stress, typically transport, lactation, food deprivation, or a combination of these factors. Characteristically the disease occurs in fat, middle-aged, pregnant, or lactating ponies that experience a decrease in feed intake. However, the disease is not restricted to this demographic, and horses or thin ponies can develop the disease. Hypertriglyceridemia is detected in horses with evidence of systemic inflammatory response syndrome (severe illness associated with decreased feed intake). There is no opacity of the plasma or serum, and the hypertriglyceridemia has not been demonstrated to worsen the outcome of the underlying disease.

PATHOGENESIS

The combination of the innate insulin resistance of ponies and a nutritional stressor, such as disease, pregnancy, lactation, or food deprivation, results in excessive mobilization of fatty acids from adipose tissue at a rate that exceeds the gluconeogenic and ketogenic capacity of the liver.1 Adipocytes of ponies, in response to norepinephrine, release fatty acids at a rate 6.5 times greater than those of horses, possibly providing at least a partial explanation for the difference in likelihood of differing breeds developing the disease. Lipolysis is mediated by β2adrenergic receptors in ponies and horses.

The induction of excessive fat mobilization in ponies is likely associated with the wellcharacterized insulin resistance of this breed, especially in obese individuals. There is no difference between ponies and horses in the extent to which lipolysis is inhibited by insulin. The effect of insulin resistance on glucose uptake from the blood might be exacerbated in sick ponies by the increase in serum cortisol concentrations associated with stress or disease. Equids have little propensity to produce ketones, and thus the excess fatty acids are reesterified in the liver to triglycerides and released into the circulation as very lowdensity lipoproteins (VLDLs). The fundamental defect in the disease is in the regulation of free fatty acid release from fat stores as a result of a defect in control of hormone-sensitive lipase, the enzyme responsible for hydrolysis of triglycerides to free fatty acids and glycerol in adipose tissue. Unchecked activity of this enzyme results in mobilization of fatty acids in hyperlipemic ponies that is 40 times the rate in normal ponies. There is no dysfunction of lipoprotein lipase, the enzyme mediating uptake of plasma free fatty acids by extrahepatic tissues, and its activity can be 300% of that of unaffected ponies. Hyperlipidemia causes widespread lipidosis and organ dysfunction. Hepatic lipidosis compromises liver function, resulting in accumulation of toxic metabolites and derangement in coagulation.

CLINICAL FINDINGS

The clinical course varies between 3 and 22 days but is generally 6 to 8 days. The unchecked disease progresses from mild depression and inappetence; through profound depression, weakness, and jaundice; to convulsions or acute death in 4 to 7 days. Depression, weight loss, and inappetence are the initial signs in 90% of cases. Approximately 50% of cases have fasciculation of muscles of the limb, trunk, or neck. Ventral edema unrelated to parturition occurs in approximately 50% of cases. Inappetence progresses to anorexia and depression, which is followed by somnolence and hepatic coma. Compulsive walking or mania develops in 30% of cases. Signs of mild colic, including flank watching, stretching, and rolling, are evident in 60% of cases. The incidence of jaundice is variable. Many animals show a willingness to drink, but they are unable to draw water into the mouth and swallow. Others continually lap at water. The temperature is normal or moderately elevated, and heart rate and respiratory rates are increased above normal. Diarrhea is an almost constant feature in the terminal stages. Visual examination of the plasma or serum phase of a blood sample collected from an affected animal reveals cloudy, milky, mildly opalescent plasma.

CLINICAL PATHOLOGY There is usually leukocytosis with neutrophilia. Hyperlipidemia is a consistent feature of the disease. Serum triglyceride concentrations will be at least 5 mmol/L (500 mg/dL) and can be much higher. Serum cholesterol and free fatty acid concentrations are also increased, although less so than triglycerides. The plasma triglyceride concentration is of minimal prognostic use in ponies, but most American miniature horses with triglyceride concentrations above 1200 mg/dL (12 mmol/L) die. Plasma glucose concentration is usually low. Ketonemia and ketonuria do not occur. Biochemical evidence of liver disease is characteristic of the advanced disease. Serum activity of gamma-glutamyltransferase (GGT) can be elevated before clinical signs of disease are apparent. Serum creatinine and urea nitrogen concentrations increase as renal function declines. Blood clotting time increases. Metabolic acidosis develops terminally. Hematologic and biochemical variables can also be affected by any underlying disease. Diagnostic confirmation of hyperlipemia is achieved by demonstration of hyperlipidemia (plasma triglyceride concentrations above 5 mmol/L [500 mg/dL]) in a horse with appropriate clinical signs. The utility of point-of-care (stall-side) analyzers has been investigated, and both units performed adequately, although not perfectly.8,9 The upper operating range of both units (~6.0 mmol/L) was lower than values for triglycerides in severely affected animals, which limits their usefulness, although it does allow identification of equids with very high concentrations. The instruments have coefficients of variation for measurement of the same sample of 10% to 16%, which limits their usefulness in monitoring responses to treatment. These analyzers are useful for field measurement of triglyceride concentrations in equids, but care should be taken in evaluating values that exceed the range of the instrument.

NECROPSY FINDINGS

Extensive fatty change is present in most internal organs, but especially in the liver, which is yellow to orange, swollen, and friable. Liver rupture with intraabdominal hemorrhage may be present. Tissue pallor as a result of lipid accumulation is also prominent in the kidney, heart, skeletal muscle, and adrenal cortex. Serosal hemorrhages of the viscera reflect disseminated intravascular coagulation. The necropsy should also include an examination for lesions that might predispose the animal to hyperlipidemia, such as pancreatic damage or laminitis. Histologically, widespread microvascular thrombosis and intracellular lipid in various tissues are evident.

Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma)

VetBooks.ir

Samples for Postmortem Confirmation of Diagnosis Samples for postmortem diagnosis include formalin-fixed liver, kidney, heart, adrenal, skeletal muscle, and pancreas for light microscopic examination. DIFFERENTIAL DIAGNOSIS • Parasitism • Anemia • Liver disease, including pyrrolizidine toxicosis • Serum hepatitis • Aflatoxicosis Hyperlipemia should be considered in any pony with a history of weight loss, inappetence, and progressive somnolence, especially in late pregnancy or early lactation.

TREATMENT • The principles of treatment are as follows: • Treatment of the underlying or inciting disease • Restoration and maintenance of a positive energy balance • Correction of any defects in hydration, acid–base, and electrolyte status • Reduction of the hyperlipidemia Every effort should be made to determine whether there is an underlying disease, and if so, it should be treated aggressively. Parasitism is a common inciting disease, as are equine Cushing’s disease and neoplasia (lymphosarcoma, gastric squamous cell carcinoma) in older ponies. The negative energy balance must be corrected. A mature, nonpregnant, and nonlactating 200-kg (440-lb) pony has energy requirements (digestible energy intake) of 9.3 Mcal/d (38 mJ/d), whereas a lactating pony has energy requirements of 13.7 Mcal/d (57.2 mJ/d). Affected animals should be encouraged to eat and must be supplemented either orally or intravenously if they will not eat a sufficient quantity. Supplements, either oral or IV, are unlikely to meet all the animal’s energy requirements, but normalization and stabilization of blood glucose concentrations, and the apparent consequent changes in hormonal milieu, inhibit lipolysis and enhance clearance of triglycerides from plasma and hepatic and renal tissues. Oral supplementation using commercial equine or human enteral nutrition preparations has been successful for treatment of the disease in American miniature horses and donkeys. If these products are not available, a homemade gruel consisting of alfalfa pellets and cottage cheese can be used. These preparations are administered every 6 hours through a nasogastric tube. Alternatively, glucose can be given orally (1 g/kg, as 5% solution every 6 hours, about 5 L to a 250-kg pony) or intravenously (5% solution,

100 mL/kg per day as a continuous IV infusion). As noted earlier, this dose of glucose will not meet the energy needs of the pony, but it might be sufficient, along with treatment of the underlying disease and supportive care, to restore normal fat metabolism. Provision of parenteral nutrition is feasible and apparently effective, but expensive and technically demanding, thereby restricting its use to veterinary hospitals.10,11 Mares in late pregnancy can be aborted, and lactating mares should have the foal removed. Dehydration and abnormalities in electrolyte and acid–base status should be corrected by oral or IV administration of isotonic fluids (lactated Ringer’s solution) and, if necessary, sodium bicarbonate. Encephalopathy associated with liver failure should be treated with oral neomycin (20 mg/kg, every 6 hours) or lactulose (1 mL/kg, every 6 hours). Hyperlipidemia should be reduced by minimizing free fatty acid production by adipose tissue and enhancing triglyceride removal from plasma. Free fatty acid production is minimized by ensuring adequate energy intake and normal plasma glucose concentrations. Use of insulin and heparin has been recommended for reduction of plasma free fatty acid concentration. However, the efficacy of these treatments is not clear, and the emphasis should be placed on provision of adequate energy intake rather than administration of these hormones. Insulin (protamine zinc insulin) is administered at 0.1 to 0.3 IU/kg SC every 12 to 24 hours. Blood glucose concentrations should be monitored, and the insulin dose may need to be adjusted. Heparin (40 to 100 IU/kg SC every 6 to 12 hours) can be given to increase lipoprotein lipase activity and promote the clearance of triglycerides from plasma. It should be noted that lipoprotein lipase activity is not deficient in affected ponies, and therefore the administration of heparin to ponies with hyperlipemia is not recommended. Severely affected ponies may have an increase in clotting time that could be exacerbated by heparin. Corticosteroids and adrenocorticotropic hormone are contraindicated in treatment of this disease.

CONTROL

A mature, nonpregnant, and nonlactating 200-kg (440-lb) pony has energy requirements of 9.3 Mcal/d (38 mJ/d), whereas a lactating pony has energy requirements of 13.7 Mcal/d (57.2 mJ/d), and every effort should be made to meet these requirements. This might require dietary supplementation during periods of nutritional stress, such as drought, late pregnancy, peak lactation, or transportation. Ponies should be maintained in optimal body condition, and nutritional stress should be avoided. A parasite and disease control program should be instituted.

1739

Transport of pregnant and lactating ponies should be avoided. FURTHER READING

Hughes KJ, et al. Equine hyperlipemia: a review. Aust Vet J. 2004;82:136. McKenzie HA. Equine hyperlipidemias. Vet Clin North Am Equine Pract. 2011;27:59.

REFERENCES 1. Radostits O, et al. Equine hyperlipemia. In: Veterinary Medicine: a Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2006:1678. 2. Burden FA, et al. J Vet Int Med. 2011;25:1420. 3. Waitt LH, et al. JAVMA. 2009;234:915. 4. Armengou L, et al. J Vet Int Med. 2013;27:567. 5. Ollivett TL, et al. Equine Vet J. 2012;(suppl):96. 6. Dunkel B, et al. Equine Vet J. 2014;46:118. 7. Oikawa S, et al. J Vet Med Sci. 2006;68:353. 8. Naylor RJ, et al. Vet Rec. 2012;170:228B. 9. Williams A, et al. Equine Vet Educ. 2012;24:520. 10. Durham AE. Vet Rec. 2006;158:159. 11. Magdesian KG. Equine Vet Educ. 2010;22:364.

Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma) Disorders of thyroid function as a result of abnormalities in the thyroid gland, pituitary gland, or hypothalamus are uncommon in the domestic species and are best documented for the horse. Thyroid disorders secondary to excessive or inadequate intake of iodine or selenium deficiency are discussed under those headings. Animals with low concentrations of thyroid hormones, usually total T3 and total T4, in blood could have nonthyroidal illness syndrome, which is well described in humans and dogs.1 Furthermore, neonates have lower concentrations of thyroid hormones in blood than do adults, and premature neonates have even lower concentrations.2

ETIOLOGY

Disorders of thyroid function result in hypothyroidism or hyperthyroidism.3 Hypothyroidism can result from diseases of the thyroid gland (primary hypothyroidism), pituitary gland (secondary hypothyroidism as a result of reduced secretion of thyroidstimulating hormone), or hypothalamus (tertiary hypothyroidism, decreased thyrotropin [thyroid-releasing hormone] secretion). Autoimmune thyroiditis has not been described in horses. Lymphocytic thyroiditis occurs in goats. Consumption of propylthiouracil (4 mg/kg body weight orally once daily for 4 to 6 weeks) induces hypothyroidism in adult horses. Administration of trimethoprim-sulfadiazine (30 mg/kg orally q24 h for 8 weeks), which can induce

1740

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

hypothyroidism in humans and dogs, does not impair thyroid function of most horses. Systemic illness, such as sepsis, or starvation can alter function of the hypothalamic–pituitary–thyroid axis, resulting in euthyroid sick syndrome, more recently termed nonthyroidal illness syndrome. The syndrome has been documented in adult horses1 and in foals with septic and nonseptic illnesses.2,4,5 Hereditary congenital hypothyroidism secondary to defects in thyroglobulin production occurs in sheep, goats, and Afrikander cattle. The disease is inherited as an autosomal-recessive trait. The cause of congenital hypothyroidism in foals is uncertain, although ingestion of nitrates by the pregnant dam is strongly suspected. Partial thyroidectomy of equine fetuses results in birth of foals with clinical and pathologic characteristics similar to those in the spontaneous disease. Hyperthyroidism in horses is attributable to functional adenocarcinoma or adenoma of the thyroid gland, but most thyroid tumors are not functional.6-8

Exercise and participation in endurance racing or competitive show jumping usually, but not always, influences serum concentrations of thyroid hormones, but is not considered a pathologic process.9,10 The response depends on the type and intensity of exercise—endurance exercise (40 to 420 km) reduces plasma concentrations of T3 and T4 at the end of the race, with return to basal concentrations by 24 hours.11 Serum T4 concentrations are lower in overtrained or malconditioned young Standardbred horses.12 Thyroid tumors are common in older horses, with ~50% having adenomas evident on histologic examination of the thyroid gland. The clinical course of such tumors is benign, although their size can be quite impressive. Thyroid adenocarcinoma is much less common but has a malignant course.6-8 Fetal undernutrition of lambs during late gestation adversely affects postnatal thyroid function and causes hyperthyroidism as adult sheep.13

EPIDEMIOLOGY

Clinical characteristics of hypothyroidism in adult horses are poorly defined, largely because of the difficulty of confirming the diagnosis and the pharmacologic effect of exogenous thyroid hormones. Clinical abnormalities anecdotally attributed to hypothyroidism include exercise intolerance, infertility, weight gain, maldistribution of body fat, agalactia, anhidrosis, and laminitis, among others. Peripheral neuropathy and keratitis sicca (secondary to facial nerve dysfunction) responsive to levothyroxine administration has been reported in a horse.14 Definitive association of these clinical syndromes with abnormalities of thyroid function is lacking. Thyroidectomy of horses causes a reduction in resting heart rate and body tem­ perature, docility, decreased food intake, increased cold sensitivity, dull hair coat, and delayed shedding of hair. Blood and plasma volumes of horses increased after removal of the thyroid glands. Effects of thyroidectomy were reversed by administration of thyroxine, with the exception of blood and plasma volume that did not return to euthyroid values. Thyroidectomized horses did not become obese or develop laminitis. Induced hypothyroidism in goats is evident as a loss of body weight, facial edema, weakness, profound depression, and loss of libido. Congenitally hypothyroid foals have a prolonged gestation but are born with a short and silky hair coat, soft and pliable ears, difficulty in standing, lax joints, and poorly ossified bones. The foals are referred to as dysmature. Characteristic musculoskeletal abnormalities include inferior (mandibular) prognathism, flexural deformities, ruptured common and lateral extensor tendons, and poorly ossified cuboidal bones.

The frequency with which hypothyroidism occurs in adult horses is unknown. It is relatively common practice to administer thyroid hormone or iodinated casein to fat horses; to those with laminitis, rhabdomyolysis, or anhidrosis; or to enhance fertility, but documentation of abnormal thyroid function in these animals is rare. None of 79 clinically normal brood mares had an abnormal response to thyroid-stimulating hormone administration, indicating that hypothyroidism is uncommon in this type of animal. Importantly, horses with nonthyroid-related illness often have low concentrations of thyroid hormones in the blood without evidence of thyroid dysfunction—this is referred to as the euthyroid sick or nonthyroidal illness syndrome and is not indicative of thyroid disease. Abnormalities of the thyroid gland were detected in 12% of 1972 goats examined in India. Of thyroid glands examined from 1000 goats in India, 2.4% had colloid goiter, 39% had parenchymatous goiter, 1.8% had lymphocytic thyroiditis, and 2.1% were fibrotic. Congenital hypothyroidism in foals occurs in western Canada and the western and northern United States. One survey of necropsy records of almost 3000 equine fetuses and neonatal foals in western Canada found that 2.7% had histologic evidence of thyroid and musculoskeletal abnormalities consistent with congenital hypothyroidism. Congenital hypothyroidism occurs in Dutch goats, Merino sheep, and Afrikander cattle. Hypothyroidism is reported in an East Friesian ram. Hyperthyroidism is a sporadic disease of older horses for which other risk factors are not identified.

CLINICAL FINDINGS

Horses with hyperthyroidism are tachycardic, display cachexia, and have hyperactive behavior. There is usually detectable enlargement of the thyroid gland both on physical examination and on scintigraphic imaging. Thyroid adenomas are evident as a unilateral nonpainful enlargement of the thyroid gland of older (>15 years) horses and are detectable on scintigraphic examination.15 Ultrasonographic and scintigraphic imaging of the thyroid gland of healthy horses is described.16 Thyroid adenocarcinoma presents as metastatic disease with both local and distant spread. Some affected horses have signs of hyperthyroidism, although this is unusual.6 Anhidrosis in horses is not associated with abnormal thyroid function.17

CLINICAL PATHOLOGY

Hematologic abnormalities in hypothyroid horses are not well documented. Induced hypothyroidism in horses causes increases in serum concentrations of VLDLs, triglycerides, and cholesterol, and decreased concentrations of NEFAs. Induced hypothyroidism in goats caused hypoglycemia, hypercholesterolemia, and anemia. Hypothyroidism in a ram caused hypercholesterolemia. Thyroid Hormone Assays Assays are available for measurement of serum concentrations of T3, T4, free T4 (fT4), free T3 (fT3; radioimmunoassay or equilibrium dialysis), and/or TSH in various species.18-20 Values of each of these analytes vary depending on the method of analysis, physiologic status of the animal, and administration of other compounds (Table 17-11). Serum concentrations of thyroid hormones are high at birth and decline with age in ruminants and nonruminants.5,21,22 For example, serum T3 concentrations of weaned Thoroughbred foals declined from 2.89 to 0.29 nmol/L at 7 and 9 months of age, serum T4 concentrations from 100.17 to 21.77 nmoL at 1 month and at 10 months, serum fT3 concentrations were 6.96 and 1.50 pmol/L at 1 month and 4 months of age, and serum fT4 concentrations were 31.40 and 4.93 pmol/L at 1 month and 9 months of age.22 There are statistically significant diurnal variations in serum concentrations of T3 and T4 in adult horses, with the lowest concentrations observed during the early morning hours, likely coincident with the time at which metabolic rate is lowest (Table 17-8). There is not a seasonal variation in thyroid hormone concentrations in horses.23 Feed restriction for 3 to 5 days lowers serum concentrations of T3, T4, and fT4 in horses by 24% to 42%. Administration of phenylbutazone decreases concentrations of fT4 (measured by equilibrium dialysis) and T4 by 4 days of treatment, which can persist for up to 10 days after discontinuation of phenylbutazone. The decrease in T4 is suggested

Disorders of Thyroid Function (Hypothyroidism, Hyperthyroidism, Congenital Hypothyroidism, Thyroid Adenoma)

1741

VetBooks.ir

Table 17-11  Serum or plasma concentrations of thyroid hormones and thyroid-stimulating hormone in foals, horses, donkeys, and cattle Physiologic status Age Birth ( 0.1) than calves with goiter that live. In the absence of TSH assay, the T4/T3 ratio can be used to diagnose hypothyroidism in newborn calves.6 Feed iodine concentration was 175 mg/kg DM (reference range > 1200 mg/kg DM) in a herd of cattle with 10% incidence of stillbirth and death of newborns as a result of iodine deficiency.9 Thyroid-weight : birth-weight ratios grea­ ter than 0.8 g/kg in newborn lambs are indicative of iodine deficiency. Ratios less than 0.4 g/kg rarely occur in lambs of flocks deficient in iodine. Intermediate ratios are ambiguous.2 The relationship between thyroid and body weight is not linear and is best defined by a probit plot (Fig. 17-10), and this nonlinear relationship should be considered when interpreting these ratios for diagnosis of iodine deficiency and need for supplementation.2 Other tests are concentrations of iodine in plasma, milk, and urine, all of which reflect current iodine status rather than revealing a profile or providing indications of previous iodine status. Estimations of iodine levels in the blood and milk are reliable indicators of the iodine status of the animal. There may be betweenbreed differences in blood iodine levels, but levels of 2.4 to 14 µg of protein-bound iodine per 100 mL of plasma appear to be in the normal range. In ewes, an iodine concentration in milk of below 8 µg/L indicates a state of iodine deficiency. Bulk-tank milk iodine content should be greater than 300 µg/L.

1746

Chapter 17 

■ 

Metabolic and Endocrine Diseases

CONTROL

VetBooks.ir

Probability that flock will respond to iodine supplementation (%)

100 90

35

0 0.1

0.4

0.8 1

10

Thyroid-weight:birth-weight ratio (g/kg) Fig. 17-10  Plot of probability that a flock of ewes will respond to iodine supplementation based on lamb thyroid-weight : body-weight ratios. A ratio of 0.40 g/kg (95% confidence interval [CI] = 0.29 to 0.47) predicted with 35% probability, and a ratio of 0.80 g/kg (95% CI = 0.70 to 0.99) predicted with 90% probability, that a lamb came from an unsupplemented (i.e., iodine-deficient) flock. (Reproduced with permission from Knowles SO, et al. N Z Vet J. 2007;55:314.)

Changes in serum thyroid hormone levels in newborn calves have been used as a diagnostic index in endemic goiter, but their high variation has been unreliable. The T4/ T3 ratio of calves with goiter was lower than in healthy calves and adult cows, and it may be a useful diagnostic aid. In determining the iodine status of an area, iodine levels in soil and pasture should be obtained, but the relationship between these levels, and between them and the status of the grazing animal, may be complicated by conditioning factors.

NECROPSY FINDINGS

Macroscopic thyroid enlargement, alopecia, and myxedema may be evident. The weights of thyroid glands have diagnostic value. In full-term normal calves the average fresh weight is 6.5 g; in lambs 2 g is average. Newborn lambs from ewes unsupplemented with iodine had a mean ratio of thyroid weight (g) to body weight (kg) of 0.40 g/kg or greater. In calves with severe thyroid hypertrophy, the gland may be heavier than 20 g. The iodine content of the thyroid will also give some indication of the iodine status of the animal. At birth, a level of 0.03% of iodine on a wet-weight basis (0.1% on dry weight) can be considered to be the critical level in cattle and sheep. On histologic examination, hyperplasia of the glandular epithelium may be seen. Follicles depleted of colloid, infolded, and lined by columnar epithelium are indicative of hypothyroidism in lambs born from ewes unsupplemented with iodine. The hair follicles will be found to be hypoplastic. Delayed osseous maturation, manifested by absence of centers of ossification, is also apparent in goitrous newborn lambs.

Samples for Confirmation of Diagnosis • Toxicology—1 thyroid gland (assay [iodine]) • Histology—skin, thyroid (LM) DIFFERENTIAL DIAGNOSIS Iodine deficiency is easily diagnosed if goiter is present, but the occurrence of stillbirths without obvious goiter may be confusing. Abortion as a result of infectious agents in cattle and sheep must be considered in these circumstances. In stillbirths resulting from iodine deficiency, gestation is usually prolonged beyond the normal period, although this may be difficult to determine in animals bred at pasture. Inherited defects of thyroid hormone synthesis are listed under the heading of inherited diseases. Hyperplastic goiter without gland enlargement has been observed in newborn foals in which rupture of the common digital extensor tendons, forelimb contracture, and mandibular prognathism also occur. The cause of the combination of defects in unknown.

TREATMENT When outbreaks of iodine deficiency occur in neonates, the emphasis is usually on providing additional iodine to the pregnant dams. The recommendations for control can be adapted to the treatment of affected animals. During an outbreak, oral administration of 280 mg/head potassium iodide to pregnant ewes and provision of iodized salt licks is advisable.1 Lambs with goiter can be administered 20 mg potassium iodide per os, once.1

The recommended dietary intake of iodine for cattle is 0.8 to 1.2 mg/kg DM of feed for lactating and pregnant cows and 0.1 to 0.3 mg/kg DM of feed for nonpregnant cows and calves. Monitoring of lamb thyroid :  body-weight ratios in areas at risk for iodine deficiency can be useful in determining the need for supplementation before and/or during pregnancy.2 Thyroid-weight : birthweight ratios greater than 0.8 g/kg are indicative of iodine deficiency, and ewes should be supplemented premating or during pregnancy to prevent goiter the following year. Ratios less than 0.4 g/kg rarely occurred among deficient flocks, so the probability of benefit from supplementation is low. Intermediate ratios are ambiguous, and individual-farm supplementation trials might be required to detect and manage the risks of marginal deficiency. Pastures in New Zealand that contain 0.24 mg iodine/kg DM provide an adequate intake for dairy cows. The injection of iodine (iodized oil) IM three times at a dose of 2370 mg iodine/dose at the start of lactation and at 100-day intervals increased iodine concentrations in milk to 58 µg/L for at least 98 days after each treatment. Two iodine injections at 100-day intervals increased milk iodine concentrations to 160 µg/L and 211 µg/L at least 55 days after each treatment, but had no effect on serum thyroid hormone concentrations. Iodine supplementation had no effect on milk, milk fat, or milk protein yield. Increasing iodine concentration in milk by IM injection of iodine could provide a method for increasing iodine intakes of humans, especially children. Iodine can be provided in salt or a mineral mixture. The loss of iodine from salt blocks may be appreciable, and an iodine preparation that is stable but contains sufficient available iodine is required. Potassium iodate satisfies these requirements and should be provided as 200 mg of potassium iodate per kilogram of salt. Potassium iodide alone is unsuitable, but when mixed with calcium stearate (8% of the stearate in potassium iodide) it is suitable for addition to salt— 200 mg/kg of salt. Individual dosing of pregnant ewes, on two occasions during the fourth and fifth months of pregnancy, with 280 mg potassium iodide or 370 mg potassium iodate has been found to be effective in the prevention of goiter in lambs when the ewes are on a heavy diet of kale. For individual animals, weekly application of tincture of iodine (4 mL for cattle; 2 mL for pigs and sheep) to the inside of the flank is also an effective preventive. The iodine can also be administered as an injection in poppy seed oil (containing 40% bound iodine): 1 mL given IM 7 to 9 weeks before lambing is sufficient to prevent severe goiter and neonatal mortality in the lambs. Control of goiter can be achieved for up to 2 years. The gestation period is also

Diseases Caused by Nutritional Deficiencies

VetBooks.ir

reduced to normal. A similar injection 3 to 5 weeks before lambing is less efficient. The administration of long-acting injectable iodine (iodized oil) at a dose of 390 mg iodine to ewes, 5 weeks premating, prevented goiter in newborn lambs from ewes fed swedes or swedes/turnips/kale as winter supplement. Administration of ~400 mg iodine per ewe increased serum iodine concentrations from 41 (standard deviation [SD] 12.2) µg iodine/L (n = 54) to 109 (SD 18.5) µg/L (n = 20; p < 0.001) at lambing ~99 days later, regardless of forage fed. High serum iodine concentrations persisted for 127 to 206 days after supplementation.2 Diet did not affect iodine concentrations in ewe serum or milk. Responses of serum total iodine concentration (an elemental determination that comprises the iodinated hormones plus various chemical forms of serum inorganic iodine7) to injection of iodized oil to sheep are proportional to dose level increasing from 42 µg/L to approximately 150 and 240 mu g/L for sheep administered either 300 mg or 400 mg of iodine, remaining elevated for 161 days.2 Milk concentrations of iodine were 26, 271, and 425 µg/L for sheep administered no supplemental iodine or 300 mg or 400 mg, respectively. Mean serum iodine concentrations of lambs from supplemented ewes with 300 mg or 400 mg iodine were 237 and 287 mu g I/L at birth, and by weaning were similar (62 ± 3 mu g/L). Concentrations in lambs born of ewes that were not supplemented were less than ~140 µg/L and were markedly affected by the diet of the ewe.7 Administration of 0.45 mg or 0.90 mg of potassium iodide orally daily to crossbred dairy goats increased mean milk iodine concentrations from 60.1 ± 50.5 (unsupplemented goats) to 78.8 ± 55.4 and 130.2 ± 62.0 mu g/L (mean ± SD), respectively. Milk production was not affected.10 A device to release iodine slowly into the forestomaches, while still retaining its position there, has given good results in preventing congenital goiter in lambs when fed to ewes during late pregnancy. A recommended approach for iodine supplementation in sheep is as follows:2 1. If feeding Brassica crops, then supplement ewes. 2. If any lamb thyroid-weight : birth-weight ratio is greater than 0.8 g/kg, then supplement ewes. The relationship between thyroid weight and body weight is not linear and is best defined by a probit plot (Fig. 17-10), and this nonlinear relationship should be considered when interpreting these ratios for diagnosis of iodine deficiency and need for supplementation.2 3. If all or most thyroid-weight : birthweight ratios are less than 0.4 g/kg, there is probably no need to supplement ewes because the probability of benefit is low.

4. If many thyroid-weight : birth-weight ratios fall between 0.4 and 0.8 g/kg, then the iodine status of the flock is unclear and can be impossible to determine from biomarkers. Supplement the ewes if other evidence is persuasive, such as occurrence of iodine deficiency in the district. An on-farm supplementation trial might be required to detect marginal deficiency on these properties. REFERENCES

1. Campbell AJD, et al. Aust Vet J. 2012;90:235. 2. Knowles SO, et al. N Z Vet J. 2007;55:314. 3. Robertson SM, et al. Aust J Exp Agr. 2008;48:995. 4. Metzner M, et al. Vet Radiol Ultrasound. 2015;56: 301. 5. Ong CB, et al. J Vet Diagn Invest. 2014;26:810. 6. Guyot H, et al. Cattle Pract. 2007;15:271. 7. Knowles SO, et al. J Anim Sci. 2015;93:425. 8. Todini L. Animal. 2007;1:997. 9. Annon. Vet Rec. 2011;169:461. 10. Nudda A, et al. J Dairy Sci. 2009;92:5133.

INHERITED GOITER Inherited goiter is recorded in Merino sheep, Afrikaner cattle, crossbred Saanen dwarf goats, Boer goats, possibly Poll Dorset sheep, and pigs, and it appears to be inherited as a recessive character. The essential defect is in the synthesis of abnormal thyroid hormone, leading to increased production of thyrotropic factor in the pituitary gland, causing in turn a hyperplasia of the thyroid gland. In Afrikaner cattle the defect stems from an abnormality of the basic RNA, and heterozygotes can be identified by blot hybridization analysis. Clinically in sheep, there is a high level of mortality, enlargement of the thyroid above the normal 2.8 g (but varying greatly up to 222 g), and the appearance of lustrous or silky wool in the fleeces of some lambs. Other defects that occur concurrently are edema and floppiness of ears, enlargement and outward or inward bowing of the front legs at the knees, and dorsoventral flattening of the nasal area. The thyroglobulin deficiency in the neonatal lamb may result in defective fetal lung development and the appearance of neonatal respiratory distress syndrome; there is dyspnea at birth. The clinical picture in goats is the same as for lambs. It includes retardation of growth, sluggish behavior, rough and sparse hair coat that worsens as the goats get older, and thick and scaly skin. In Afrikaner cattle, most of the losses are from stillbirths or from early neonatal deaths. Some are caused by tracheal compression from the enlarged gland. It is the calves with the largest glands that have the greatest mortality. In these cattle there may be a concurrent inherited gray coat color, a defect in a red breed. In pigs, hairless and swollen piglets with enlarged thyroid glands occur, in the

1747

proportions with normal piglets consistent with an autosomal-recessive mode of inheritance.

Diseases Caused by Nutritional Deficiencies INTRODUCTION Three criteria are suggested for the assessment of the importance of nutrition in the etiology of a disease state in a single animal or in a group of animals: • Is there evidence from an examination of the diet that a deficiency of a specific nutrient or nutrients may be occurring? • Is there evidence from an examination of the animals that a deficiency of the suspected essential nutrient or nutrients could cause the observed disease? • Does supplementation of the diet with the essential nutrient or nutrients prevent or cure the condition? The difficulties encountered in satisfying these criteria, and making an accurate and reliable diagnosis of a nutritional deficiency, increase as investigations progress into the area of trace element and vitamin nutrition. The concentration of these micronutrients in feedstuffs and body tissues are exceedingly small, and assays are often difficult and expensive. Because of these difficulties it is becoming more acceptable to describe individual syndromes as “responsive diseases”— that is, the investigation satisfies only the third of the previously listed three criteria. This practice is not ideal, but has advantages in that it is more a more cost-effective approach, and relevant control measures are directly assessed.

EVIDENCE OF A DEFICIENCY AS THE CAUSE OF THE DISEASE Evidence of a deficiency as the cause of the disease will include evidence of a deficiency in the diet or an abnormal absorption, utilization, or requirement of the nutrient under consideration. Additional evidence may be obtained by chemical or biological examination of the feed. Diet The diet for a considerable period before the occurrence of the disease must be considered because body stores of most dietary factors may delay the appearance of clinical signs. Specific deficiencies are likely to be associated with particular soil types, and in many instances national or local soil and geological maps may predict the likely occurrence of a nutritional disease. Diseases of plants may also indicate specific soil deficiencies, such as “reclamation disease” of oats, which indicates copper deficiency. The predominant

1748

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

plant species in the pasture sward may also be important; subterranean clover selectively absorbs copper, legumes selectively absorb molybdenum, and Astragalus spp. accumulate selenium. Farming practices can have a strong influence on the concentration of specific nutrients in livestock feed. For example, heavy applications of nitrogen fertilizer can reduce the copper, cobalt, molybdenum, and manganese content of pasture. On the other hand, many applications of lime will reduce the concentration of copper, cobalt, zinc, and manganese in plants, but increase molybdenum. These effects are significant enough to influence the trace-element nutrition of grazing livestock. Modern hay-making methods, with their emphasis on the artificial drying of immature forage, tend to conserve vitamin A, but may result in a gross deficiency of vitamin D. Improved pasture species and increased applications of fertilizer can exaggerate the depletion of trace elements from marginally deficient soil, giving rise to overt deficiency disease in previously marginal or unaffected areas. Thus local knowledge of farming and feeding practices in a particular area is of primary importance in the diagnosis of nutritional deficiency states. Abnormal Absorption Although a diet may contain adequate amounts of a particular nutrient, some other factor, by decreasing its absorption, may induce a deficiency. For example, excess phosphate reduces calcium absorption, excess calcium reduces the absorption of iodine, and the absence of bile salts prevents proper absorption of the fat-soluble vitamins. Chronic enteritis reduces the absorption of most essential nutrients. The list of antagonisms that exist between elements continues to grow, most being an interference with absorption. For example, excess calcium in the diet interferes with the absorption of fluorine, lead, zinc, and cadmium, such that it may cause nutritional deficiencies of these elements, but it also reduces their toxic effects when they are present in the diet in excessive amounts. Abnormal Utilization of Ingested Nutrients Abnormal utilization of ingested nutrients may also have an effect on the development of conditioned deficiency diseases. For example, molybdenum and sulfate reduce copper storage, vitamin E has a sparing effect on vitamin A, and thiamine reduces the dietary requirement for essential fatty acids. Abnormal Requirement An enhanced growth rate of animals, either by improved nutrition or genetic selection, may increase their requirement for specific nutrients to the point where deficiency disease occurs. There seems to be little doubt that there is a genetic variation in mineral

metabolism, and it has been suggested that it may be possible to breed sheep to “fit” deficiency conditions. The significance of the inherited component of an animal’s nutritional requirement is unknown, but should not be overlooked when policies to upgrade livestock in deficient areas are being planned.

EVIDENCE OF A DEFICIENCY ASSOCIATED WITH THE DISEASE Evidence of a deficiency associated with the disease is usually available from experimental work that demonstrates the clinical signs and necropsy findings produced by each deficiency. Several modifying factors may confuse the issue. Under natural circumstances, nutritional deficiencies may not be a single entity, and thus clinical and necropsy findings will often be complicated by deficiencies of other factors and intercurrent infections. Most syndromes are variable and insidious in their onset, and clinical signs and gross necropsy lesions in many nutritional deficiency diseases are either minimal or nonspecific. This increases the challenge of making a definitive diagnosis. Consequently, laboratory examination of blood and animal tissues is an essential diagnostic aid in many instances. However, the normal range of blood or tissue concentrations of minerals and vitamins, or their biochemical markers, and those values that indicate deficiency, are often not well established. Experimentally induced and naturally occurring nutritional deficiencies provide an indication of the changes that occur in the concentrations of a particular nutrient, but variations resulting from age, genotype, production cycle, length of time on the inadequate diet, previous body stores of the element, and intercurrent disease and stressors can complicate the results, making them difficult to interpret. In most cases, nutritional deficiencies affect a proportion of the herd or the flock at the same time. The clinicopathological examination should include a selection of both normal and clinically affected animals because the comparison of results from these groups allows a more accurate and reliable interpretation of laboratory tests, facilitating a diagnosis.

EVIDENCE BASED ON CURE OR PREVENTION BY CORRECTION OF THE DEFICIENCY The best test of the diagnosis of a suspected nutritional deficiency is to observe the effect of supplementing that nutrient, either directly to the animal or via the ration. Confounding factors can occur, such as spontaneous recovery; hence, adequate controls and a sufficient sample size are essential. Curative responses may be poor because of an inadequate dose or advanced tissue damage. Alternatively, the abnormality may

have only been a predisposing or secondary factor to another factor that is still present. A common cause of confounding in therapeutic trials is the impurity or bioactivity of the preparations used, particularly with trace elements and vitamins. The preparations used may also have some intrinsic pharmacologic activity and hence partially or temporarily ameliorate the condition without a deficiency actually having been present. Monitoring of Nutritional Status On breeding farms, there are several different age groups of animals at different levels of growth and production. This requires close surveillance to avoid either a deficiency or overnutrition in each class of animal. Scoring of the body condition of dairy and beef cattle, sheep, and pigs is commonly used as an indicator of the adequacy of the diet leading to the present time (termed prior nutrition). The feeds and feeding program have a major influence on reproductive performance, and hence growth and milk production and must be monitored regularly. The veterinarian must be aware of any changes in the feeding program that have occurred since the last farm visit or that are intended in the near future. Veterinary clinical nutrition is now a specialty that should provide new and useful information for the practitioner working with a particular species or class of food animals. An experienced and competent nutritionist should be consulted to assist with complex nutritional problems. Nutritional Management in Dairy Herds Advising farms about nutrition is a key activity for dairy cattle practitioners. Feed costs are approximately 60% of the total cost of producing milk, so even minor improvements in feeding efficiency can be profitable. Some dairy practitioners function as the nutritional specialist for the dairy farms they serve, collecting feed samples for nutrient analysis, formulating rations, and advising on crop and harvesting conditions. These veterinarians often devote a considerable amount of their professional time to nutritional management. Nevertheless, it is common for farms to employ a professional nutritionist or to use a nutritionist employed by a feed company or local cooperative. These professionals generally formulate the rations and submit feed samples for nutrient analysis. For these herds, the veterinarian can have an important role in ensuring that the diet described on paper is adequately formulated and delivered to the cows. Routine scheduled activities, such as measuring the dry matter of forages, hand-mixing of total mixed ration (TMR) for one cow and comparing it with the machine-mixed TMR (termed the TMR test mix), and scoring the feed bunk to assess feed sorting and dry matter intake are important procedures that

Diseases Caused by Nutritional Deficiencies

VetBooks.ir

help to ensure the successful delivery of a nutritional program. Assessing pasture conditions by periodic inspection of pasture is an important component of managing the nutritional program of herds that use management-intensive grazing. These quality control activities should be conducted routinely as part of the health and production management program. There is probably no aspect of a dairy enterprise that has a wider impact than the feeding program, which has direct effects on production and growth. Many health problems on a dairy farm relate in some way to the feeding program, and a significant portion of the farm’s labor is spent planting, growing, harvesting, mixing, and feeding rations. Investment in equipment used in feeding programs is also an important capital cost. Small changes in feeding programs may bring about large changes in productivity, health, income, feed costs, labor allocation, cash flow, and debt. Thus the total savings from these small changes can be substantial, with one study showing that routine nutritional consultation by a veterinarian can save 14% of total feed costs even without accounting for improved production or health effects. For these reasons, veterinarians who wish to serve their dairy clients on a whole-farm basis must become actively involved in the herd’s feeding program. Dairy herds are often fed unbalanced, expensive rations, but as a consultant independent from the feed company, a veterinarian can provide unbiased advice about the feed program. For example, a recumbent, hypocalcemic cow raises questions about dry-cow feeding, whereas an anestrous, thin cow with smooth ovaries raises questions about energy and dry matter intake (DMI) during early lactation. If the average mature equivalent milk production for the herd falls by 220 kg (500 lb), this generates the same sense of urgency as a cow with a prolapsed uterus. A dairy veterinarian cannot truly serve a client’s needs by practicing therapeutic medicine in isolation from the nutrition of the herd and thus must acquire skills to directly deal with nutrition problems. As the average size of dairy herds increases, many dairy farmers now rely on a team of advisors rather than just one or two. Consequently, a nutritional consultant, local veterinarian, and remote specialist consultant may all be providing advice to a dairy farm, and thus awareness of and communication about the feeding program, and the indicators of performance of the farm, are critical. It imperative that, as part of the advisory team, the dairy veterinarian knows about dairy nutrition and is aware of, but preferably involved in, the ration formulation. Levels of Nutritional Service Having decided to be involved in a dairy’s feeding program, a first step is for the client and veterinarian to discuss and agree on

the level of nutritional advice that is to be provided. This varies from herd to herd, depending on the veterinarian’s expertise, the client’s ability and interest, and the role of other consultants. There are essentially four levels of service that might be provided, as described next. Level 1: Problem Identification and Analysis At level 1, the veterinarian takes on the task of monitoring the dairy herd for indicators of nutrition-related problems. Many areas need to be monitored: production, milk composition, DMI, body scores, disease rates, heifer management and growth, and feed costs. Based on these measures, the veterinarian can identify problems as they arise, form and test hypotheses about likely causes, and interact with the client and other advisors as the problems are prioritized and addressed. Level 2: Ration Analysis Level 2 requires assessment of the adequacy of diets that are actually being fed to the cows. Problems of balance or economics are referred to the appropriate person, for example, if reformulation of a specific diet is needed. This involvement may be difficult to sustain if the person formulating the ration resents being “second-guessed,” but it can work well if a functional team approach is in place. Level 3: Ration Formulation If a dairy veterinarian takes responsibility for ration formulation, the veterinarian will need considerably enhanced skills in dairy nutrition, far beyond those traditionally taught at veterinary colleges. Typically, this involves using a computer program to formulate a balanced, least-cost ration for each class of animal. It requires expertise in the mechanics of how feeds are handled and fed to cows on a daily basis, and hence an intimate knowledge of the farm and its personnel and daily trends in the price and availability of feed components. If not well managed, this level of service has several pitfalls because it lacks the on-farm follow-up, supervision of implementation, and monitoring of results that are included in level 4. There is a truism about feeding dairy cows that every cow has three rations: the one formulated, the one delivered, and the one actually eaten. The best feeding programs minimize the difference among these three rations. If the veterinary consultant’s role stops at formulation, then mistakes can occur in delivery and feedbunk management that can doom the program to failure. However, if the program fails, it is the ration formulation that is most likely to be blamed. Level 4: Total Program Consulting Level 4 service includes the critical aspects missing from level 3 because the veterinarian

1749

plays an active role in implementing the feeding recommendations. Attention is paid to areas such as bunk management, cow comfort, feeding frequency and scheduling, quality control, and consistency of feeding management. Working closely with the producer, plans for future forage production can be generated, including attention to factors such as timing the harvest for maximum feed value. The monitoring described in level 1 is sustained, and timely adjustments and feedback are provided to ensure that the rations are accomplishing the desired ends. In the long term, this is the level of service that is most desirable for both the specialist dairy veterinarian and client. The producer benefits from the added supervision and support, and the veterinarian can assure the client that the program is implemented as it is intended. If it is not working, the total program can be modified, often with the veterinarian as a part of a team that includes a nutritionist. When multiple consultants are used in larger herds, this team approach provides the owner with the best opportunity for expert advice, and the specialist dairy veterinarian is often best suited to be the “team leader.” Nutritional Management of the Beef Breeding Herd Good nutrition provides the essential basis for optimum productivity in cattle-breeding operations. Despite this, nutritional expertise has not been a traditional strength of many food-animal veterinarians. Throughout the world, beef-breeding operations are generally range or pasture based. These operations are conducted in diverse environments, with great variation in nutritional management. In many countries, the area of pasture or rangeland required to maintain a cow–calf unit may vary from 0.5 to 1 ha (1 or 2 acres) in intensive high-rainfall regions to many square kilometers in remote dryland areas. However, in general, the land area or amount of pasture necessary for production is related to local economic realities. This, in turn, is related to levels of managerial and resource inputs that differ markedly between regions, markets, and enterprises. Notwithstanding this variation, there are a number of principles of good nutritional management that can be universally applied to cattle-breeding operations. Regardless of region, an important consideration is that of maintaining or improving production (increasing income) while reducing costs per unit of production. In simple terms, financial return from a beef-breeding operation is a function of number of calves, their weaning weight, and price. On the cost side is the cost of maintaining the breeding females. This varies considerably between farms, both within and between regions. Although it can be influenced, the price received is generally not significantly controlled by the farm business. However, both

1750

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

the number of calves born and their weaning weights are strongly influenced by an appropriate management calendar that matches nutritional demand with the supply of pasture. For example, good nutritional management helps to ensure that as many females as possible are cycling at the start of the breeding season. This, combined with good bull management, helps to ensure that calves are born early and that they are older and heavier at weaning than later-born calves. In general, nutrition is the most important limiting factor of beef-breeding performance, and thus an understanding of the principles underlying the nutritional management of breeding females is essential. Effective monitoring does not necessarily require a higher degree in nutrition, although it should include sufficient knowledge and wisdom to know when additional expertise is needed. A starting point is to have a working knowledge of the different energy measuring systems (total digestible nutrients [TDN], metabolizable energy [ME], and net energy, NE) that are commonly used and their applications for different classes of animals, activities, and feedstuffs, and to identify one with which the veterinarian can work best. The Nutrient Requirements of Beef Cattle from the National Research Council (NRC) in the United States is a useful document, with the 7th edition published in 2000. This is packaged with a computer program that includes ration formulators and a library of feeds and feedstuffs. A number of programs for least-cost-ration formulation in beef herds are also available from Departments of Agriculture or commercial suppliers. Nutritional Advice for Beef Feedlots Beef feedlots frequently consult a qualified nutritionist to assist in the formulation of cost-effective rations. In this case the veterinarian should communicate regularly with the nutritionist to be aware of the composition of the diets and any changes that are planned. Because feed is the major portion of the cost per unit of body weight gain, it is imperative that the diet be the lowest-cost diet possible while providing nutrients that allow optimum growth and finishing. Most of the emphasis in feedlot nutrition has been on the development of cost-effective diets that support a maximum growth rate without any deleterious effects. Considerable information is available on the nutrient requirements for feedlot cattle and on the feeds and feeding systems used. The precise specifications of the diets are the responsibility of the nutritionist, but the feedlot veterinarian is often able to evaluate the quality of the feed delivery system. This includes whether cattle are fed on time, whether the feed delivered to troughs is properly mixed, and whether feed intake is intermittent as a result of insufficient trough space, poor trough and pen design,

inclement weather, and muddy or slippery ground. Any deviations should be discussed with feedlot managers and the consulting nutritionist, similar to the team approach suggested for large dairy herds. Nutritional deficiency diseases are uncommon in feedlot cattle because cattle usually receive a diet that contains the nutrients required for maintenance and pro­ motion of rapid growth. Diets prepared according to the Nutrient Requirements of Beef Cattle should meet all the requirements under most conditions. Specific nutrient deficiencies are extremely rare because diets are prepared every few days or daily, and it would be highly unusual for a feedlot to use a feedstuff deficient in a specific nutrient for a prolonged period. However, such a situation may occur on a small farm or opportunistic feedlot that prepares its own diet with little or no attention to the need to supplement homegrown feeds. Thus there are only a few nutrition-related diseases that may affect a well-managed feedlot, but these diseases may cause large economic losses when they occur. They include the following: • Carbohydrate engorgement (grain overload or lactic acidosis) • Feedlot bloat or ruminal tympany • Feeding errors, including accidental incorporation of an excessive amount of feed additives, such as monensin or urea; sudden unintended changes in the composition of the diet; and accidental feeding of the wrong dietary mix Nutritional Advice for Swine-Herds Veterinarians involved in health management of swine-herds must be well informed about the nutrient requirements of the different age groups of pigs. Feed constitutes 60% to 80% of the cost of producing a market pig, so every effort is needed to increase the efficiency of feed use. Surveys of well-managed pig farms in Alberta, Canada, found a 20% difference in feed costs, and it is estimated that in the industry the range in feed costs is likely to be near 50%. Reduction of the feed cost of the highest-costing farm to that of the lowest-costing farm would save that farm more than US$23,000 annually, a reduction in the cost of production of $6.80/pig. The trend is to use complete feeds formulated by feed company nutritionists familiar with the nutrient composition of local feedstuffs. With complete diets, specific nutrient deficiencies are uncommon. The major problem is the efficiency of utilization of the different feeds throughout the life cycle of the pig. The nutrient requirements of the pig at various phases of growth, from birth to market weight and of breeding stock, are well established. The remaining questions relate to the amount of feed provided during the different growth phases of the pig to achieve optimum production and yield the best carcass. The following are some

recommended practices for increasing efficiency of feed utilization: • Provide well-balanced diets with adequate levels of amino acids, energy, vitamins, and minerals necessary to meet the particular demands of the pig at each stage of its life cycle. The diet depends on the demands, usually characterized as the growth rate or lean deposition, with feed intake being the supply function. Feed intake is limited by appetite, and thus other nutrients are matched to expected energy intake and subsequent growth. • Use least-cost formulation to the extent that it is feasible. The least-cost energy source in most of the pig-rearing areas is corn, and the most common protein source is soybean meal. • Restrict the level of a properly balanced diet for sows during gestation to avoid overfeeding. Sows that have lost excessive body weight in the previous lactation need supplemental feed during the dry period to avoid thin-sow syndrome. • Ad-lib feeding for growing pigs is usually optimal unless the genotype deposits excess fat during the latter stages of growth. • Market pigs as close to optimum slaughter weight as possible to maximize margin over feed costs. • Avoid feed wastage by using welldesigned feeding systems and proper adjustment of feeders. • Use pelleting of diets to increase digestibility, especially of small grains, and to decrease feed wastage. However, pelleting does predispose pigs to gastroesophageal ulcers. The feed efficiency of the pigs from weaning to market should be monitored regularly. It is often difficult to obtain accurate data for a specific group of pigs because a common feeding system for multiple groups is often used. However, the total amount of feed used and the total weight of pigs marketed will give an estimate of feed efficiency. Although the nutrient requirements of pigs are well known, they do continue to change because of changes in growth and production characteristics. Pigs with high lean-growth rates require higher levels of amino acids to support their increased rate of body protein deposition. Similarly, highmilk-producing sows nursing large litters have increased amino acid requirements. The NRC in the United States provides an important service in establishing the nutrient requirements of swine and other species. The 10th edition of the Nutrient Requirements of Swine was published in 1998, and it includes areas such as modeling nutrient requirements and reducing nutrient excretion, particularly nitrogen and phosphorus, which can contribute to environmental pollution. The approach used to produce estimates of nutrient requirements account for the

Diseases Caused by Nutritional Deficiencies

VetBooks.ir

pig’s body weight and the accretion of lean (protein) tissue, gender, health status, and various environmental factors. To accurately estimate nutrient needs of gestating and lactating sows, there is a need to account for body weight, weight gain during gestation, weight loss during lactation, number of pigs in the litter, weight gain of the litter (a reflection of milk yield), and certain environmental factors. A series of integrated equations is used to account for the many factors known to influence nutrient requirements. These provide the framework for modeling the biological basis of predicting requirements. The NRC models predict the levels of nutrients (outputs) needed to achieve a certain level of production under a given set of environmental conditions (inputs). Five principles were used to develop the models: (1) ease of use by people with varying levels of nutritional expertise; (2) continued relevance; (3) structural simplicity; (4) transparency, so that all equations are available to the user; and (5) empirical data at the whole-animal level was used rather than data based on theoretical values. Three independent models were developed for growth, gestation, and lactation. The growth model estimates amino acid requirements of pigs from weaning to market weight, and the gestation and lactation models estimate energy and amino acid requirements of gestating and lactating sows. Few revisions were needed from the previously published mineral requirements; higher dietary requirements for sodium and chloride in the young pig were established, and manganese requirements were increased from 10 to 20 ppm for gestating and lactating sows. Feed composition tables are built from multiple databases on the nutrient composition of feeds, including the feed industry and datasets outside the United States and Canada. The information on water was expanded, with more detailed information on the factors that influence water intake. Information on nonnutritive feed additives, such as antimicrobial agents, anthelmintics, microbial supplements, oligosaccharides, enzymes, acidifiers, flavors, odor-control agents, antioxidant pellet binders, flow agents, highmineral supplements, and carcass modifiers, is also included. Nutritional Advice for Sheep Flocks The influence of nutrition on the reproductive performance of ewes has been a matter of concern for many years. Clearly, the relationship between the provision of nutrients and requirements for optimum reproductive performance is seldom ideal because of the wide range of environmental conditions and the seasonal breeding patterns of most sheep breeds. Prolonged periods of undernutrition often occur during midpregnancy, partly the result of the decline in feed availability and

quality over that stage of the reproductive cycle and partly from the seasonal variability in pasture growth. Prolonged moderate to severe undernutrition of ewes bearing twins in midpregnancy reduces placental development and can cause a significant reduction in lamb birth weight and increased lamb mortalities. Considerable progress has been made in understanding the principles of nutrition of sheep and in defining their nutrient requirements for maintenance, pregnancy, and lactation. It has been established that mortality rates are high in lambs with birth weights below the breed norm, and that after birth the absolute growth rates are lower in surviving light lambs than in heavier lambs of the same breed. The plane of nutrition and the size of the placenta have been recognized as major determinants of the fetal growth rate. Fetal growth retardation in undernourished ewes has a placental component, and thus factors that affect placental growth are highly relevant. The 21-week gestation can be divided into a number of periods to consider the effects of nutrition on reproduction within each period. In the first 4 weeks of gestation, embryonic loss is the main sequelae of inadequate nutrition. During this period, it is generally recommended that the bodycondition score (BCS) of the ewe be maintained at an average of 3.0, on a scale of 1 (emaciated) to 5 (very fat), to minimize embryonic and early fetal loss. This is followed by a period of 2 months in which there is rapid growth of the placenta, but during which growth of the fetus in absolute terms is still small. Over this period, losses in body weight should not exceed 5%, and BCS should be maintained at 2.7 to 3.0. Finally, there is the phase from 90 days to parturition, in which gain in the mass of the fetus amounts to 85% of its birth weight, during which time nutrient intake must be increased if excessive weight loss in the ewe and lightbirth-weight lambs are to be avoided. Placental and Fetal Growth Placental development in the pregnant ewe begins about 30 days after conception. The number of placentomes associated with each fetus is fixed at this time, but the total weight of the placentomes increases until about 90 days of gestation, after which there is little change. The factors that influence the ultimate size of the placenta and its weight include hormonal and nutritional factors, prolonged environmental heating of pregnant ewes, parity, and possibly genotype. However, by far the most important determinant is nutrition of the ewe. Moderately severe undernutrition during early and midpregnancy significantly reduces placental weight at term and causes chronic intrauterine growth retardation. The size of the placenta is a major determinant of fetal growth. In well-fed ewes, the

1751

fetal growth rate until 120 days (17 weeks) of gestation is not positively correlated with placental weight, but fetal growth rate is limited by the size of the placenta during the last 3 to 4 weeks of pregnancy. However, when ewes are underfed, the influences of a lighter placenta on fetal growth rate are evident sooner, with placental weight and fetal growth positively correlated from as early as 90 days (13 weeks) of gestation. During the first 90 days of pregnancy, placental growth is reduced when ewes are moderately underfed. Light fetuses in ewes with placenta weights near the bottom of the normal range suffer chronic and progressive hypoxemia and hypoglycemia. This affects fetal metabolism, causing fetal death during late pregnancy, fetal hypoxemia during parturition, premature birth, and a high perinatal mortality rate from hypoglycemia and hypothermia, the latter being more severe in lighter lambs. The extent to which ewes maintained on a fixed ration draw on their own body reserves in an attempt to meet the energy costs of pregnancy is determined by fetal weight. In well-fed ewes, fetal growth rate remains constant until at least 120 days of gestation and then decreases. However, the absolute growth rate increases markedly during the last 8 weeks of gestation, when fetal growth is most rapid, exceeding 100 g/ day near birth. The growth rate among fetuses is highly variable, which accounts for birth weights ranging from 2 kg to over 7 kg. When ewes that have previously been well fed are severely underfed at any stage during the last 40 to 50 days of pregnancy, fetal growth rate decreases by 30% to 70% within 3 days. This demonstrates that mobilization of maternal reserves is substantially less than fetal requirements, emphasizing the importance of a continuous supply of good-quality feed during late pregnancy. The larger the fetal burden, the more susceptible an ewe is to hypoglycemia during underfeeding. Refeeding after severe underfeeding can reverse the reduced growth rate of fetuses, but the response depends on the duration of the underfeeding. If the period of underfeeding is 16 days or less, the growth rate increases when ewes are refed, but there is no change when refeeding occurs after 21 days of severe underfeeding. Moderate underfeeding of pregnant ewes for 85 days reduces the fetal growth rate irreversibly. Refeeding them in late pregnancy does not cause fetal growth rate to increase, but it does prevent further decreases after 120 days. Lamb Losses The major consequences of prenatal growth retardation are on lamb survival. Neonatal mortality increases markedly in many environments when the birth weight falls below 3 to 3.5 kg. Compared with normal lambs, low-birth-weight animals have reduced insulation because of the smaller number of wool

1752

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

fibers, greater relative heat loss because of their larger surface area per unit of body weight, and a reduced capability to maintain heat production because of their lower fat and energy reserves. All of these factors increase their susceptibility to environmental stress and reduce their ability to compete with normal-sized siblings. Underfeeding during pregnancy reduces available body lipids in lambs by about 47%, and it also decreases the lactose, lipid, and protein available in colostrum during the first 18 hours after birth by about 50%. Newborn lambs have to draw on body reserves of glycogen to maintain heat production during the first 18 hours after birth. Consequently, they depend heavily on colostrum and supplements, when these are provided, to avoid hypoglycemia and hypothermia. The effects of maternal nutrition on udder development and on the production and yield of colostrum and milk in ewes have also been examined. In the 30 days before birth, there is a marked increase in the rate of mammary tissue growth in the ewe. In well-fed ewes with one or two lambs, large volumes of colostrum accumulate in the mammary glands during the last few days of pregnancy, and copious milk secretion begins soon after birth, with averages of 1800 to 2800 mL of colostrum and milk being produced during the first 18 hours. Udder growth rates show a similar pattern to fetal growth rates, such that the greatest increase in udder weight occurs in the last 30 days of gestation, and the weight of udder tissue is 30% to 40% of the total weight of the litter. Colostrum production is proportional to udder weight, but refeeding ewes a few days before lambing fills the udder tissue present rather than increasing udder tissue weight. In underfed ewes, accumulation of colostrum before birth is reduced markedly, lactogenesis is delayed, and the total production of colostrum and milk during the first 18 h averages only 1000 mL. Subsequently, for ewes on both planes of nutrition, milk production increases, reaching a peak about 1 to 2 weeks after birth. Underfeeding ewes from 105 days (15 weeks) of gestation can reduce the total yield of colostrum during the first 18 hours after birth by decreasing mammary tissue growth. Thus the prepartum accumulation of colostrum and its subsequent rates of secretion are reduced. Improving the ewe’s nutrition from 1 hour after birth can increase the secretion rates of colostrum between 10 and 18 hours. The growth rate of lambs during the first few weeks of life is positively correlated with birth weight. Low planes of maternal nutrition during late pregnancy and early lactation are generally associated with low birth weights, milk yields, and postnatal growth rates, and high planes of nutrition are associated with the opposite effects. A marked increase in the plane of maternal nutrition at birth can overcome the inhibitory effects on

lactation and lamb growth rate of underfeeding in late pregnancy. Ewe Body-Condition Score Target condition scores for ewes at different stages of their reproductive cycle have been developed by research groups and departments of agriculture in many countries. These vary according to the predominant breeds and production systems in each country, and they can be quite different for a Merino, Dorset, or Friesian ewe used for wool, meat, or dairy production versus a dual-purpose enterprise producing both meat and wool. Consequently, readers should directly access information appropriate to the production systems of their clients. However, in general, the aim at breeding time is to have ewes with a BCS of 3.0 to 3.5, which ensures maximum ovulation rate. Ewes with a BCS of 3.5 at breeding can be allowed to lose no more than 5% of their body weight, steadily, during the second and third months of pregnancy, equivalent to approximately 0.5 to 1 BCS units. This mild degree of undernutrition enables good placental growth, establishing the basis for maximum fetal growth in the fourth and fifth months of pregnancy, during which the fetus achieves over 80% of its growth. During these final 2 months of pregnancy, there is a limit to the extent to which body-fat reserves can be used because excessive mobilization of fat deposits as a consequence of inadequate dietary energy supply leads to pregnancy toxemia. Ewes with a BCS below 3.0 should be managed to maintain that score. In late gestation, the optimum BCS ranges from 2.75 to 3.0. In contrast, early lactation is a period in which body fat can be safely used to meet some of the high-energy demands of lactation. During this period, a loss of BCS of from 0.5 to 1.0 (equivalent to 5 kg of fat for a 70-kg ewe at mating) may occur. However, replacement of body fat, to increase the BCS to 3.0 to 3.5 before the next mating, is important to maximize ovulation rate and achieve optimum reproductive performance. Winter shearing of pregnant ewes during the final 10 weeks of pregnancy can cause a significant increase in lamb birth weight by stimulating ewe appetite. However, this also increases the base energy requirements of the ewe at a feed-limiting time of the year in many production systems (e.g., winter for a spring-lambing flock). Thus it is not always an optimum or profitable system, but this will vary between production systems and different countries. The nutrient requirements for maintenance, breeding, pregnancy, and lactation of ewes have been cataloged, and optimum feeding strategies for the breeding ewe can be formulated. The evaluation of the ewes’ ration during late gestation by monitoring plasma concentrations of the BHB has been described, and these evaluations have been

used to provide nutritional advice in intensively managed flocks during late gestation. In more intensively managed flocks, achieving optimum reproductive performance requires adjusting feeding strategies and the nutrient value of the diet to meet the needs of each stage of the reproductive cycle. Requirement for metabolizable energy increases above maintenance levels from 8 to 12 weeks of pregnancy, increasing further in late pregnancy and lactation. During early lactation, when the energy requirements of prolific ewes exceed the voluntary intake from all but the highest-quality diets, bodyfat reserves are used and then replenished toward the end of lactation, when milk yield declines, and in the period leading up to rebreeding. The rapid growth of the fetus after 90 days of pregnancy and increased energy demand may require the feeding of cereal or legume concentrates, rather than hay, which has far lower metabolizable energy content. This is particularly true for ewes carrying twins or triplets. In contrast to the ability of the ewe to use body reserves when the intake of energy fails to meet her needs, particularly in early lactation, there is little scope for sustaining production by drawing on body protein. For example, lactating ewes can lose up to 7 kg of body fat during a 4-week period in early lactation, when energy intake is below requirements. For ewes on a low-protein intake, the maximum daily loss of protein is around 26 g. Therefore it is important to meet the protein needs of the ewe during pregnancy, but especially during late pregnancy, to ensure adequate fetal growth, udder development, and colostrum production. The estimates for the minimum protein requirements of the animal are based on distinguishing between the needs of the rumen microflora for rumen-degradable protein and of the host animal for additional undegraded dietary protein when rumen-degradable protein fails to meet those requirements. In practice, the dietary allowances for late pregnancy and early lactation are higher than the sum of the rumen-degradable protein and undegradable protein. FURTHER READING Freer M, ed. Nutrient Requirements of Domesticated Ruminants [eBook]. Melbourne: CSIRO Publishing; 2007. Freer M, Dove H, eds. Sheep Nutrition. Wallingford, Oxon, UK: CSIRO and CABI Publishing; 2002. Hayton A, Husband J, Vecqueray R. Nutritional management of herd health. In: Green M, ed. [eBook]. Dairy Herd Health. Wallingford, Oxon, UK: CAB International; 2012. Herring AD. Beef Cattle Production Systems. Wallingford, Oxon, UK: CAB International; 2014. Subcommittee on Dairy Cattle Nutrition, Committee on Animal Nutrition, Board on Agriculture, National Research Council Subcommittee on Dairy Cattle Nutrition. Nutrient Requirements of Dairy Cattle. 7th ed. Washington, DC: National Academy Press; 2000.

Deficiencies of Energy and Protein

VetBooks.ir

Subcommittee on Beef Cattle Nutrition, Committee on Animal Nutrition, Board on Agriculture, National Research Council. Nutrient Requirements of Beef Cattle. 7th rev. ed. Washington, DC: National Academy Press; 2000. Subcommittee on Horse Nutrition, Committee on Animal Nutrition, Board on Agriculture, National Research Council. Nutrient Requirements of Horses. 6th rev. ed. Washington, DC: National Academy Press; 2007. Subcommittee on Sheep Nutrition, Committee on Animal Nutrition, Board on Agriculture, National Research Council. Nutrient Requirements of Sheep. 6th rev. ed. Washington, DC: National Academy Press; 1985. Subcommittee on Swine Nutrition, Committee on Animal Nutrition, Board on Agriculture, National Research Council. Nutrient Requirements of Swine. 10th rev. ed. Washington, DC: National Academy Press; 1998.

Deficiencies of Energy and Protein DEFICIENCY OF ENERGY ETIOLOGY Insufficient quantity or quality of feed is a common nutritional deficiency and practical problem of feeding livestock. The term protein-energy malnutrition is used to describe a form of incomplete starvation in which a suboptimal amount of energy and protein is present in the diet. Such deficiencies typically occur when livestock are underfed, and often the two scenarios cannot be separated.

EPIDEMIOLOGY

A deficiency of energy is the most common production-limiting nutrient deficiency of farm animals. There may be inadequate amounts of feed available, or the feed may be of low quality (low digestibility). The availability of pasture may be inadequate because of overgrazing, drought, or snow covering. Alternatively, it may be too expensive to provide enough supplementary feed of the required quality, or the available feed may be of such low quality and poor digestibility that animals cannot consume enough to meet energy requirements. In some cases, forage may contain a high concentration of water, which limits total energy intake.

CLINICAL FINDINGS

The clinical findings of an energy deficiency depend on the age of the animal, whether or not it is pregnant or lactating, concurrent deficiencies of other nutrients, and environmental factors. In general, an insufficient supply of energy in young livestock causes decreased growth and delayed onset of puberty. In mature animals, there is reduced milk production and a shortened lactation. A prolonged energy deficiency in pregnant beef heifers will result in a failure to produce adequate quantities of colostrum at parturition.

In mature animals, there is also a marked loss of body weight, especially when demand for energy increases in late pregnancy and early lactation. There are prolonged periods of anestrus, which reduces the reproductive performance of the herd. Primigravid females are particularly susceptible to protein-energy malnutrition because of their requirements for growth and maintenance. A deficiency of energy during late gestation can produce undersized, weak neonates with a high mortality rate, whereas abomasal impaction is associated with energy deficiency during prolonged cold weather, especially in pregnant beef cattle and ewes being wintered on poor-quality roughage. Heat loss from the animal to the environment increases considerably during cold weather, and when ambient temperatures are below the critical temperatures, the animal responds by increasing metabolic rate to maintain normal body core temperature.1 If sufficient feed is available when temperatures are below the lower critical temperature, ruminants will increase their voluntary feed intake to maintain body temperature. If sufficient feed is not available, the animal will mobilize energy stored as fat or muscle to maintain body temperature and thus lose body weight. In the case of ruminants and horses, if the feed is of poor quality, for example, poor-quality roughage, the increased feed intake may result in impaction of the abomasum and forestomaches in cattle and of the large intestine in the horse. Cold, windy, and wet weather will increase the needs for energy, and the effects of a deficiency are exaggerated, often resulting in weakness, recumbency, and death. A sudden dietary deficiency of energy in fat, pregnant beef cattle and ewes can result in starvation ketosis and pregnancy toxemia. Hyperlipemia occurs in fat, pregnant or lactating ponies that are on a falling plane of nutrition. Protein-energy malnutrition occurs in neonatal calves fed inferior-quality milk replacers that may contain insufficient energy or added nonmilk proteins, which may be indigestible by the newborn calf. A major portion of the body fat present at birth can be depleted in diarrheic calves deprived of milk and fed only fluids and electrolytes for 4 to 7 days. Feeding only fluids and electrolytes to normal, healthy newborn calves for 7 days can result in a significant loss of perirenal and bone-marrow fat and depletion of visible omental, mesenteric, and subcutaneous fat stores. The amount of body fat present in a calf at birth is an important determinant of the length of time an apparently healthy calf can survive in the face of malnutrition. Calves born from dams on an adequate diet usually have sufficient body fat to provide energy for at least 7 days of severe malnutrition. The absence of perirenal fat in a calf at 2 to 4 days of age suggests inadequate reserves of fat at birth and chronic fetal malnutrition.

1753

DEFICIENCY OF PROTEIN A deficiency of protein commonly accompanies a deficiency of energy. However, the effects of the protein deficiency, at least in the early stages, are usually not as severe as those of energy deficiency. Insufficient protein intake in young animals results in reduced appetite, lowered feed intake, inferior growth rate, lack of muscle development, and a prolonged time to reach maturity. In mature animals, there is loss of weight and decreased milk production. In both young and mature animals, there is a drop in hemoglobin concentration, packed cell volume, total serum protein, and serum albumin. In the late stages, there is edema associated with the hypoproteinemia. Ruminants do not normally need a dietary supply of essential amino acids, in contrast to pigs, which need a natural protein supplement in addition to the major portion of total protein supplied by the cereal grains. The amino acid composition of the dietary protein for ruminants is not critical because the ruminal flora synthesize the necessary amino acids from lowerquality proteins and nonprotein sources of nitrogen.

CLINICAL FINDINGS

The clinical findings of a protein deficiency are similar to those of an energy deficiency, and the clinical findings of both resemble those of many other specific nutrient deficiencies and subclinical diseases. Proteinenergy malnutrition in beef cattle occurs most commonly in late gestation and is characterized clinically by weakness, clinical recumbency, marked loss of body weight, a normal mental attitude, and a desire to eat. Cows with concurrent hypocalcemia will be anorexic. If the condition occurs at the time of parturition, there will be an obvious lack of colostrum. Calves of these cows may attempt to vigorously suck their dams, attempt to eat dry feed, drink surface water or urine, and bellow continuously. Affected cows and their calves may die within 7 to 10 days. Protein-energy malnutrition is less common in dairy cattle because they are usually fed to meet the requirements of maintenance and milk production. Dairy calves fed inferior-quality milk replacers during periods of cold weather will lose weight, become inactive and lethargic, and may die within 2 to 4 weeks. Affected calves may maintain their appetites until just before death. Diarrhea may occur concurrently and be confused with acute undifferentiated diarrhea as a result of the enteropathogenic viruses or cryptosporidiosis. Affected calves recover quickly when fed cow’s whole milk. Protein-energy malnutrition also occurs in sheep and, less commonly, in goats. Excessive dental attrition is a common cause in grazing sheep, which is exacerbated by the excessive ingestion of soil.

1754

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

DIFFERENTIAL DIAGNOSIS The diagnosis will depend on an estimation of the concentration of energy and protein in the feed, or a feed analysis, and comparing the results with the estimated nutrient requirements of the class of affected animals. In some cases, a sample of feed used several weeks earlier may no longer be available, or the daily feed intake may not be known. Marginal deficiencies of energy and protein may be detectable with the aid of a metabolic profile test. Specific treatment of livestock affected with protein-energy malnutrition is usually not undertaken because of the high cost and prolonged recovery period. Oral and parenteral fluid and electrolyte therapy can be given as indicated. The provision of highquality feeds appropriate to the species is the most cost-effective strategy.

PREVENTION The prevention of protein-energy malnutrition requires the provision of the nutrient requirements of the animals according to age, stage of pregnancy and production, the environmental temperature, and the cost of the feeds. Body-condition scoring of cattle and sheep can be used as a guide to monitor body condition and nutritional status. Regular analysis of feed supplies will assist in the overall nutritional management program. The published nutrient requirements of domestic animals are only guidelines to estimated requirements because they were determined in experimental animals selected for uniform size and other characteristics. Under practical conditions, all of the common factors that affect requirements must be considered. FURTHER READING Freer M, ed. Nutrient Requirements of Domesticated Ruminants [eBook]. Melbourne: CSIRO Publishing; 2007. Freer M, Dove H, eds. Sheep Nutrition. Wallingford, Oxon, UK: CSIRO and CABI Publishing; 2002. Hayton A, Husband J, Vecqueray R. Nutritional management of herd health. In: Green M, ed. [eBook]. Dairy Herd Health. Wallingford, Oxon, UK: CAB International; 2012. Herring AD. Beef Cattle Production Systems. Wallingford, Oxon, UK: CAB International; 2014.

REFERENCE

1. Grazfeed v 5.04, CSIRO. Accessed at ; June 18, 2016.

LOW-MILK-FAT SYNDROME In low-milk-fat syndrome, the concentration of fat in milk is reduced, often to less than 50% of normal, while milk volume is maintained. This syndrome is a significant cause of wastage in high-producing cows. Low concentration of fat in milk occurs with ruminal acidosis in cattle.1 The cause appears to be an increase in concentrations

of conjugated linoleic acid in the diet, with subsequent reduction in lipogenesis in the udder.2 A supply of polyunsaturated fatty acids in the cows’ ration and alteration in fermentation in the rumen results in biohydrogenation of linoleic acid (abundant in oils and seeds) and formation of intermediate fatty acids in the rumen. These incompletely hydrogenated fatty acids are absorbed into the blood and have an inhibitory effect on lipogenesis.3 This syndrome occurs most commonly in cows on low-fiber diets, for example, lush, irrigated pasture or grain rations that are ground very finely or fed as pellets. Treatment is achieved by administration of sodium bicarbonate or magnesium oxide, which increase fiber digestibility and hence the propionate : acetate ratio. Magnesium oxide also increases the activity of lipoprotein lipase in the mammary gland and increases uptake of triglycerides by the mammary gland from the plasma.4 REFERENCES

1. Atkinson O. Cattle Pract. 2014;22:1. 2. Gulati SK, et al. Can J Anim Sci. 2006;86:63. 3. Dubuc J, et al. Point Veterinaire. 2009;40:45. 4. Radostits O, et al. Veterinary Medicine: a Textbook of the Diseases of Cattle, Horses, Sheep, Goats, and Pigs. 10th ed. London: W.B. Saunders; 2006: 1686.

Diseases Associated With Deficiencies of Mineral Nutrients There is an enormous literature about mineral nutrient deficiencies in livestock, and thus it is not possible to comprehensively review it here, but some general comments are appropriate. In developed countries, severe deficiencies of single elements affecting very large numbers of animals now seldom occur. The diagnostic research work has been done, the guidelines for preventive programs have been outlined, and these have been applied in the field. Thus the major contributions to knowledge have already been made, and what remains is essentially applying and extending that knowledge. Some loose ends remain, including preventing the overzealous or unnecessary application of minerals, which can produce toxicoses or is simply not cost-effective; sorting out the relative importance of the constituent elements in combined deficiencies, characterized by incomplete response to single elements; and devising better ways of detecting marginal deficiencies. At least 15 mineral elements are essential nutrients for ruminants. The macrominerals, required daily in gram amounts, are calcium, phosphorous, potassium, sodium, chlorine, magnesium, and sulfur. The trace elements, or microminerals, are copper, selenium, zinc, cobalt, iron, iodine, manganese, and

molybdenum. Improving trace-element nutrition of grazing livestock, in a way that is cost-effective and that meets consumer perceptions and preferences, is a continuing challenge.1

PREVALENCE AND ECONOMIC IMPORTANCE Despite experimental evidence that deficiencies or excesses of trace elements can influence growth, reproductive performance, or immunocompetence of livestock, there is often a lack of information on the prevalence and economic significance of such problems. Most published reports of trace-elementrelated diseases are case reports and thus provide insufficient information to assess prevalence and economic impact on a regional or national scale. Many reports are also compromised by commercial bias. Despite this, Food and Agriculture Organization/World Health Organization (FAO/ WHO) Animal Health Yearbooks show that of the countries providing information on animal diseases, 80% report nutritional diseases of moderate or high incidence, and trace-element deficiencies or toxicities are involved in more than half of those whose causes were identified. As a specific example, in the United Kingdom it has been estimated that despite the activities of its nutritional and veterinary advisory services, and extensive supplementation, clinical signs of copper deficiency occur annually in approximately 1% of the cattle population. Copper deficiency can also predispose to increased mortality as a result of infectious diseases in lambs, and so it is likely that the economic losses from copper deficiency may be considerably underestimated even in developed agricultural economies.

DIAGNOSTIC STRATEGIES In developed countries with more advanced livestock industries, the emphasis is on disease prevention rather than therapy, and the cost-effective control of trace-element deficiencies is a matter of ongoing farmer education rather than research. Copper, cobalt, selenium, and iodine deficiencies can affect reproductive performance, appetite, early postnatal growth, and immunocompetence on a herd or flock basis, and thus emphasis is placed on identifying the risk of deficiency before clinical signs appear. Monitoring the trace-element status of livestock is typically done by blood, saliva, or tissue analysis, or less commonly by measuring the concentration of the trace element in the diet. An alternative way of monitoring preclinical stages of a trace element deficiency is to identify and measure a biochemical indicator that reflects changes in the activity of an enzyme involved in a key metabolic pathway, such as vitamin B12 or glutathione peroxidase, which are indirect

Diseases Associated With Deficiencies of Mineral Nutrients

VetBooks.ir

measures of cobalt and selenium nutrition in sheep, respectively. To be useful, techniques should be able to predict the likely pathologic outcome of different suboptimal concentrations of a particular measure, and hence when it is clinically and economically justifiable to apply treatments or interventions. For example, a high proportion of grazing cattle become hypocupremic if maintained on pasture forage, but they don’t develop clinical signs of deficiency, and only a small percentage exhibit any physiologic response to the administration of copper. This illustrates the variation in the development of clinical signs of copper deficiency, which can be induced by a simple dietary deficiency or by interactions between copper and other elements in the diet, such as molybdenum, sulfur, and iron. There is also evidence that genetic variation influences the utilization of trace elements by livestock. For example, there are differences in dietary requirements for copper between some breeds of sheep. Sheep can also be selected for a high or low concentration of plasma copper, which can have profound physiologic consequences in the low-copper group. Thus although it is known from soil maps and local knowledge where trace-element deficiencies occur, their prevalence and importance may be underestimated because subclinical deficiency may go unnoticed for prolonged periods.

DEFICIENCIES IN DEVELOPING COUNTRIES

clinical disease will also be influenced by their age, physiologic status (pregnant, lactating, or dry), genetic differences, and interactions with other trace elements. For example, there is good evidence that whereas dietary copper may be adequate for some breeds of sheep, such concentrations may be deficient, or even toxic, for others. Dietary deficiency does not inevitably lead to clinical disease, but several factors interact and predispose the animal to clinical disease, including the following: • Age—for example, late-term fetal lambs are highly susceptible to demyelination as a result of copper deficiency, which produces “swayback.” • Genetic differences and individual variation in response to deficiency. • Fluctuating demand for trace elements because of changes in growth, physiologic status (especially lactation), and diet. • Substitution—the use of alternative metabolic pathways in response to a deficiency, such as selenium, which may incompletely protect sheep from white-muscle disease when the diet is deficient in vitamin E. • Size of the functional reserves.

1755

The trace elements are component parts of many tissues and are often involved in metabolic pathways, either as a single key enzyme or in many interacting components. Consequently, their deficiency leads to a variety of pathologic consequences, metabolic defects, and clinical signs. These are summarized in Table 17-12. The soil and its parent materials are the primary sources of trace elements from which soil–plant–animal relationships are built. Soil maps created from geochemical surveys can help identify areas in which livestock are exposed to excessive ingestion or deficiencies of trace elements. Variations in the concentration of most trace elements in soils are quite wide, ranging from soils that are grossly deficient to those that are potentially toxic. The availability of trace elements to plants is controlled by their total concentration in the soil and their chemical form. Certain species of plants take up more trace elements than others, and the ingestion of soil can also have a profound effect on the nutrition and metabolism of some trace elements. It is often difficult to determine the role of individual trace elements in deficiency states because many trace-element

Table 17-12  Principal pathologic and metabolic defects in essential trace-element deficiencies

In developing countries, deficiencies of trace elements are often hidden or confounded by gross nutritional deficiencies of energy, protein, phosphorus, and water, which affect postnatal growth and reproductive performance. Undernutrition is the most important limitation to herbivore livestock production in tropical countries, but mineral deficiencies or imbalances in soils and pasture forages, particularly of phosphorus, cobalt, or copper, are also responsible for poor reproductive performance and low growth rates.

Deficiency

Pathologic consequence

Associated metabolic defect

Copper

Defective melanin production Defective keratinization; hair, wool Defective cross linkages in connective tissue, osteoporosis Ataxia, myelin aplasia Growth failure

Tyrosine/DOPA oxidation –SH oxidation to S–S Lysyl oxidase

Cobalt

Anorexia Impaired oxidation of propionate Anemia

Methyl malonyl CoA mutase Tetrahydrofolate methyl transferase

PATHOPHYSIOLOGY OF TRACE-ELEMENT DEFICIENCY

Selenium

Myopathy; cardiac/skeletal Liver necrosis Defective neutrophil function

Peroxide/hydroperoxide destruction Decreased glutathione peroxidase OH; O2 generation

Zinc

Anorexia, growth failure

Multifactorial; increased expression of leptin (satiety signal) and cholecystokinin (appetite regulation), reduced pyruvate kinase Polynucleotide synthesis, transcription, translation?

The physiologic basis of trace-element deficiency is complex.1 Some trace elements are essential for the function of a single enzyme, whereas others are involved in multiple metabolic pathways. Consequently, a deficiency of a specific element may affect one or more metabolic processes and produce a variety of clinical signs in different classes of livestock. Furthermore, there is a wide variation in the clinical response to decreased blood or tissue concentrations of a trace element between individuals. For example, two animals in a herd or flock with the same concentration of copper in their blood may be in different body condition. Their susceptibility to

Anemia Uricemia

Parakeratosis

Cytochrome c oxidase Decreased biogenic molecules such as gastrin Ceruloplasmin (ferroxidase) Urate oxidase

Perinatal mortality Thymic involution Defective cell-mediated immunity Iodine

Thyroid hyperplasia Reproductive failure Hair, wool loss

Decreased thyroid hormone synthesis

Manganese

Skeletal/cartilage defects Reproductive failure

Chondroitin sulfate synthesis

Chapter 17 

■ 

Metabolic and Endocrine Diseases

VetBooks.ir

deficiencies produce nonspecific and specific clinical signs, especially when complex interactions occur. Consequently, the dose– response trial still has a significant role to investigate complex or marginal deficiencies and whether a cost-effective response will occur on a particular farm.2 A properly conducted dose–response trial requires comparison of the response to treatment, typically a biochemical indicator and a measure of production, such as body weight, in a supplemented and control group. Ideally, animals should be randomly selected and allocated to groups, and the groups should be of sufficient size to reliably detect an economically significant difference (e.g., have a 95% chance of detecting a 1-kg difference in body weight). Where appropriate, the control (unsupplemented) group should be treated with the vehicle or inactive portion of the substance given to the supplemented group (a placebo). Additional requirements for a reliable dose–response trial include a careful appraisal of the reasons for conducting the trial, a suitable form of treatment, and a reliable biochemical method for monitoring the response to the trace element. Dose–response trials establish a link between a trace element and certain clinical signs. They can also identify factors that modify the response to a trace element and, importantly, provide some indication of the economic response to supplementation. The ad hoc field observations made by veterinarians who make a diagnosis of a trace-element deficiency, followed by treatment or dietary changes, are subjective and usually lack controls. Nevertheless, they are useful in that they indicate the magnitude and variability of response that might be expected in future studies. There are major challenges in predicting and diagnosing trace-element deficiencies in grazing livestock, including complex interactions between dietary constituents and the homeostatic mechanisms of the animal. Thus it is usually impossible to predict from the composition or analysis of the diet whether clinical signs of deficiency will occur. Consequently, assessment of the absorbable, rather than the total, concentration of elements in the diet is now considered to be more important in understanding the nutritional basis for the deficiencies, but tests of the livestock are a more definitive assessment of deficiency.

LABORATORY DIAGNOSIS OF MINERAL DEFICIENCIES The diagnosis of mineral deficiencies, particularly trace-element deficiencies, relies heavily on the interpretation of the bio­ chemical tests. This is because deficiencies of any one or more of several trace elements can result in nonspecific clinical abnormalities, such as loss of weight, growth retardation, anorexia, and inferior reproductive performance.

100

1. Storage

2. Transport 3. Function

Marginal supply

Nutrient pool

1756

4. Clinical signs

0

Depletion

Deficiency

Dysfunction

Disease

Time Fig. 17-11  The sequence of pathophysiological changes that can occur in mineral-deprived livestock, commencing with depletion and ending with clinical disease, and their relation to the body pools of that nutrient. (Reproduced with permission from Suttle NF. Mineral Nutrition of Livestock. 4th ed. Wallingford, Oxon, UK: CAB International; 2010 [Chapter 1].)

The interpretation of biochemical criteria of trace-element status is governed by three important principles: relationship with intake, time, and function. These are further explained as follows: 1. Relationship between the tissue concentrations of a direct marker and the dietary intake of the element will generally be sigmoid in shape (a dose–response curve). The important point on the curve is the intake at which the requirement of the animal is passed, which is the intake of the nutrient needed to maintain normal physiologic concentrations of the element and/or avoid impairment of essential functions. For several markers of trace-element status, the position on the x-axis at which the requirement is passed coincides with the end of the lower plateau of the response in marker concentration. Under these conditions, the marker is an excellent index of sufficiency and body reserves, but an insensitive index of a deficiency. If requirement is passed at the beginning of the upper plateau, the marker is a poor index of sufficiency, but a good index of deficiency. This principle allows direct markers to be divided into storage and nonstorage types corresponding to the former and latter positions on the x-axis. 2. Nonstorage criteria can be divided into indicators of acute and chronic deficiency, and two types of relationships can be distinguished: a rapid, early decline in marker concentration followed by a plateau; and a slow, linear rate of decline. Markers with a slow, linear response will be good indices of a chronic deficiency, but unreliable indices of acute deficiency, because they cannot respond quickly

enough. Conversely, the marker with a rapid, early decline will be a good index of acute deficiency, but an unreliable indicator for chronic deficiency if the low plateau is reached before functions are impaired. Those biochemical criteria that are based on metalloenzyme or metalloprotein concentrations in erythrocytes are of the slow type because the marker is incorporated into the erythrocyte before its release into the bloodstream, and thereafter its half-life is determined by that of the erythrocyte, which is 150 days or more. Metalloenzymes or metalloproteins in the plasma with short half-lives provide markers of the rapid type. 3. A deficiency can be divided into four phases: depletion, deficiency (marginal), dysfunction, and clinical disease. During these phases there are progressive changes in the body pools of mineral that serve as storage (e.g., liver for copper, bone for Ca and P), transport (e.g., plasma), and function (e.g., muscle enzymes) (Fig. 17-11).3 Depletion is a relative term describing the failure of the diet to maintain the traceelement status of the body, and it may continue for weeks or months without observable clinical effects when substantial body reserves exist. When the net requirement for an essential element exceeds the net flow of the absorbed element across the intestine, then depletion occurs. The body processes may respond by improving intestinal absorption or decreasing endogenous losses. During the depletion phase, there is a loss of trace element from any storage sites, such as the liver, during which time the plasma concentrations of the trace element may remain constant. The liver is a common store for copper, iron, and vitamins A and B12.

Diseases Associated With Deficiencies of Mineral Nutrients

VetBooks.ir

If the dietary deficiency persists, eventually there is a transition from a state of depletion to one of deficiency, which is marked by biochemical indications that the homeostatic mechanisms are no longer maintaining a constant level of trace elements necessary for normal physiologic function. After variable periods of time, the concentrations or activities of trace-element-containing enzymes will begin to decline, leading to the phase of dysfunction. There may be a further lag period, the subclinical phase, before the changes in cellular function are manifested as clinical disease. The biochemical criteria can be divided, according to the phase during which they change, into indicators of marginal deficiency and dysfunction. The rate of onset of clinical disease will depend on the intensity of the dietary deficiency, the duration of the deficit, and the size of the initial

reserve. If reserves are nonexistent, as with zinc metabolism, the effects may be acute, and the separate phases become superimposed. The application of these principles to the interpretation of biochemical criteria of trace-element status is presented elsewhere where applicable, in the discussion of each mineral nutrient. The definitive etiologic diagnosis of a trace-element deficiency will depend on the response in growth and health obtained following treatment or supplementation of the diet. The concurrent measurement of biochemical markers will aid in the interpretation and validation of those markers for future diagnosis. The strategies for anticipating and preventing trace-element deficiencies include regular analysis of the feed and soil, which is not highly reliable, and monitoring samples from herds and flocks to

1757

prevent animals from entering the zone of marginal trace-element deficiencies that precedes the onset of functional deficiency. The decision to intervene can be safely based on the conventional criteria of marginal traceelement status. FURTHER READING Lee J, Masters DG, White CL, Grace ND, Judson GJ. Current issues in trace element nutrition of grazing livestock in Australia and New Zealand. Aust J Agric Res. 1999;50:1341-1364. Suttle NF. Mineral Nutrition of Livestock. 4th ed. Wallingford, Oxon, UK: CAB International; 2010.

REFERENCES

1. Suttle NF. Mineral Nutrition of Livestock. 4th ed. Wallingford, Oxon, UK: CAB International; 2010 [Chapter 1]. 2. Ibid., [Chapter 19]. 3. Ibid., [Chapter 3].

VetBooks.ir

18 

Diseases Primarily Affecting the Reproductive System

INFECTIOUS DISEASES PRIMARILY AFFECTING THE REPRODUCTIVE SYSTEM  1758 Induction of Parturition  1758 Freemartinism in Calves  1760 Buller Steer Syndrome  1760 INFECTIOUS DISEASES PRIMARILY AFFECTING THE REPRODUCTIVE SYSTEM  1761 Brucellosis Associated With Brucella abortus (Bang’s Disease)  1761 Brucellosis Associated With Brucella ovis  1774 Brucellosis Associated With Brucella suis in Pigs  1778 Brucellosis Associated With Brucella melitensis  1781 Abortion in Ewes Associated With Salmonella abortusovis  1784 Abortion in Mares and Septicemia in Foals Associated With Salmonella

Infectious Diseases Primarily Affecting the Reproductive System This chapter presents information related to important and selected livestock pathogens that affect not only the fertility but also the health of animals, and in some cases, such as bovine brucellosis, the health of humans. It is worth noting that national control and eradication campaigns against Brucella abortus infection in cattle played, and continue to play, an important global role in expanding the veterinary profession. More recently, the worldwide spread of porcine reproductive and respiratory syndrome (PRRS) virus during the last 20 years has had a marked economic impact on the swine industry. As a consequence, PRRS is currently one of the most intensively researched diseases of livestock. Readers seeking detailed information related to reproductive performance, the estrous cycle, conception, pregnancy, and parturition are directed to the many excellent textbooks that address these subjects.

INDUCTION OF PARTURITION CALVES

The induction of parturition in pregnant cows during the last 6 weeks of gestation by Copyright © 2017 Elsevier Ltd. All Rights Reserved. 1758

abortusequi (abortivoequina) (Equine Paratyphoid)  1785 Chlamydial Abortion (Enzootic Abortion of Ewes, Ovine Enzootic Abortion)  1786 Coxiellosis (Q-Fever)  1791 Diseases of the Genital Tract Associated With Mycoplasma spp.  1793 Equine Coital Exanthema  1794 Porcine Reproductive and Respiratory Syndrome (PRRS)  1794 Menangle  1816 Japanese Encephalitis (JE; Japanese B Encephalitis)  1817 Neosporosis  1817 Dourine (Maladie du coit)  1819 TOXIC AGENTS PRIMARILY AFFECTING THE REPRODUCTIVE SYSTEM  1821 Estrogenic Substances  1821 Phytoestrogen Toxicosis  1822

Zearalenone Toxicosis  1824 Mare Reproductive Loss Syndrome (Early Fetal Loss, Late Fetal Loss, Fibrinous Pericarditis, and Unilateral Uveitis)  1825 Equine Amnionitis and Fetal Loss  1827 Plants and Fungi (Unknown Toxins) Affecting the Reproductive System  1827 CONGENITAL AND INHERITED DISEASES PRIMARILY AFFECTING THE REPRODUCTIVE SYSTEM  1828 Chromosomal Translocations in Cattle  1828 Inherited Prolonged Gestation (Adenohypophyseal Hypoplasia)  1828 Inherited Inguinal Hernia and Cryptorchidism  1829

the parenteral injection of corticosteroid with or without prostaglandin F2α (PGF2α) has raised the question of animal welfare and of the possible effects of prematurity on the disease resistance of the newborn calf. The induction of premature parturition in cattle has found application in the following areas: • With pastoral-based dairy production, synchronization of the calving period has allowed maximal utilization of seasonally available pastures by the synchronization of peak demand for dry matter intake with spring flush in pasture growth. In pastoral-based herds with breeding for seasonal calving, late-calving cows will be induced and these average approximately 8% of the herd. • Ensuring that calving coincides with the availability of labor to facilitate observations and management of calving and to overcome the inconvenience caused by late-calving cows. • Minimizing dystocia in small heifers and animals with exceedingly long gestation periods (past due). • The therapeutic termination of pregnancy for various clinical reasons. • As an aid in the control of milk fever in combination with parenteral administration of vitamin D analogs. A variety of short-acting and long-acting corticosteroids have been used. A single injection of a short-acting formulation is

used when it is desirable to induce calving within the last 2 weeks of gestation. Earlier in pregnancy short-acting corticosteroids were found to be insufficiently reliable to induce parturition, which has led to the common use of long-acting corticosteroid formulations. A variety of protocols to induce premature parturition (3–6 weeks before due date) are used in practice; the main issue is the poor predictability of the time of calving relative to treatment when using long-acting corticosteroids. Common protocols use a second treatment with shortacting corticosteroids or the administration of PGF2α 50 to 10 days after the initial treatment. The use of PGF2α at least 9 days after treatment with long-acting corticosteroids was found to reliably narrow down the calving time, with the great majority of all cows calving within 72 hours of PGF2α treatment.1 The use of PGF2α did not improve the viability of the premature neonates or their survival rate. For cattle near term (within 2 weeks of due date) the use of short-acting cortico­ steroid formulation is more appropriate with parturition generally occurring within 2 to 4 days posttreatment.2 The mortality rate of induced calves is considerable and can exceed 30%, particularly when dams are induced at or before the eighth month of gestation.2 Mortality in calves born as a result of induced parturition is primarily a result of prematurity, and

Infectious Diseases Primarily Affecting the Reproductive System

VetBooks.ir

calf mortality is generally low when calving is induced within 12 days of parturition, although there are welfare concerns. The calves born earlier in pregnancy after using long-acting corticosteroid are usually lighter in weight, lethargic, and slow to stand and to suck properly. The serum immunoglobulin concentration was found to be lower in calves born from dams induced with longacting corticosteroids because of interference with intestinal absorption by the corticosteroid. Up to 60% of calves born following induction with long-acting corticosteroids are at risk for failure of transfer of passive immunity. The colostrum available to such calves also has a reduced immunoglobulin content, and there may also be a reduction in the total volume of colostrum available from the induced-calving cows. Immunoglobulin absorption rates were not impaired when short-acting corticosteroids are used to induce calving close to term. Artificial induction of parturition is an important risk factor for retention of the placenta, and the incidence is reported to vary from 20% to 100%. Subsequent reproductive performance of induced cows can be impaired. A risk for acute gram-negative bacterial infections is reported in a low (0.3%) proportion of cows following induction with dexamethasone. The use of longacting corticosteroids was also associated with a higher incidence of photosensitization in treated heifers.2 In a study where partus induction was systematically used in cows that exceeded a gestation length of 282 days, no detrimental effects on calf viability, cow health, and productive and reproductive performance during lactation were found compared with untreated control animals. The incidence of retained fetal membranes in untreated animals was not recorded in this study and could thus not be compared with treated animals.2 When parturition is induced in large herds of beef cattle, particularly with a high percentage of heifers, increased surveillance will be necessary after the calves are born to avoid mismothering. Every attempt must be made to establish the cow–calf pair (neonatal bond) and move them out of the main calving area. Heifers that disown their calves must be confined in a small pen and be encouraged to accept the calf and let it suck, which is sometimes a very unrewarding chore. Calf mortality can be very high where calving is induced earlier than 35 weeks of pregnancy.

LAMBS

The induction of parturition in sheep is not a common practice, but it can be used to synchronize lambing in flocks where there are accurate dates of mating for individual ewes. Unless accurate dates are available, there is risk of prematurity. Also, ewes that are more than 10 days from their normal parturition date are unlikely to respond.

Induction of parturition is also used as a therapeutic ploy to terminate pregnancies in sheep with pregnancy toxemia. Induction is usually performed with dexamethasone and less commonly with betamethasone or flumethasone, which is more expensive. Lambing occurs 36 to 48 hours later, and there may be breed differences in response. Variability in lambing time can be reduced by the use of clenbuterol and oxytocin.

FOALS

The induction of parturition in mares for reasons of economy, management convenience, concern for prolonged gestation, or clinical conditions such as prepubic tendon rupture or research and teaching is now being practiced. Foaling can be induced with oxytocin, ideally administered as an intravenous (IV) drip over 15 to 30 minutes, and occurs within 15 to 90 minutes of its administration. High doses of oxytocin are potentially dangerous to the foal and low doses (10–20 IU) are preferred. Glucocorticoids, and antiprogestagens that are effective in inducing pregnancy in other species, are either ineffective in the mare or capricious in their efficacy and can also be associated with adverse effects on the foal. Prostaglandin F2α and its analogs have been used for partus induction in the mare and low doses (5–12 mg intramuscularly [IM]) may be effective at term, but repeated treatments may be required. The time interval between treatment and delivery is difficult to predict and can range from 1 to 48 hours. The use of PGF2α for partus induction in mares has been discouraged because considerable risks such as premature placental separation and foal death that have been associated with this treatment.3 Induction of parturition in the mare is not without risk and has been associated with the birth of foals that are weak, injured, or susceptible to perinatal infections. The period of fetal maturation is relatively short in the horse and is considered to be the last 2 to 3 days’ gestation. Because spontaneous parturition in healthy mares can occur between 320 and 360 days, there is the risk of delivering a foal that is premature and nonviable. Fetal maturity is the major prerequisite for successful induced parturition, and the three essential criteria are • A gestational length of more than 330 days • Substantial mammary development and the presence of colostrum in the mammary gland with a calcium concentration greater than 10 mmol/L • Softening of the cervix The rise in calcium concentration is the most reliable predictor of fetal maturity and milk calcium concentrations above 10 mmol/L, in combination with a concentration of potassium that is greater than sodium, are indicative of fetal maturity. Commercial milk test strips are available for estimating mammary

1759

secretion electrolyte concentrations; however, it is recommended that testing be done in an accredited laboratory. In mature foals, head lifting, sternal recumbency, and evidence of suck reflex occurs within 5 minutes of spontaneous fullterm deliveries. The foal can stand within 1 hour and suck the mare within 2 hours. The behavior and viability of the premature foal after induced parturition have been described. The overall survival rate of foals delivered from induced parturition before 320 days’ gestation was 5%. Four patterns of neonatal adaptation were observed on the basis of righting, sucking, and standing ability. If the suck reflex was weak or absent and the foals were unable to establish righting reflexes, the prognosis of survival was poor. Foals born before 300 days’ gestation did not survive for more than 90 minutes; foals born closer to 320 days’ gestation had a better chance of survival and exhibited behavioral patterns of adaptation. In addition to the potential delivery of a premature or weak foal, other adverse effects of induction can be dystocia, premature placental separation, and retained placenta.

PIGLETS

The induction of parturition of gilts and sows on days 112, 113, or 114 of gestation is highly reliable and can be achieved by a single IM injection of 175 µg of cloprostenol or 5 to 10 mg of PGF2α. The sows farrow approximately 20 to 36 hours later. Synchronization of farrowing can be improved by administration of oxytocin (5–30 IU) 20 to 24 hours after injection of PGF2. Induction of parturition has been used on large-scale farms to allow a concentration of labor, to improve supervision and care at the time of farrowing, to reduce the incidence of the mastitis/metritis/agalactia syndrome, and to reduce the percentage of stillborn piglets. The end day of a batch farrowing system can be fixed and weekend farrowing avoided. The subsequent fertility of the sows is not impaired. Induction on day 110 may be associated with a slight increase in perinatal mortality. TREATMENT Premature partus induction cattle (>2 weeks before due date): Dexamethasone trimethyl-acetate (or other long-acting formulation) (25–30 mg/animal IM as single dose) (R-1) Dinoprost (or other PGF2α-analogon) (25 mg/ animal IM as a single dose 5–10 days after dexamethasone treatment) (R-2) Partus induction cattle (95° C, 203° F), and disinfect with formaldehyde-based product; allow disinfectant water to remain in pits overnight

2

Pump out pits, repeat washing procedure, and disinfect in phenol-based product; allow disinfectant to remain in pits

311

Allow facility to remain vacant

12

Pump out slurry pits, repeat washing procedure, and disinfect with formaldehyde-based product

13

Allow facility to remain vacant

14

Resume conventional flow of pigs into clean nurseries

weight of marketable pigs, as a result of their increased growth rate and decreased mortality. Lower treatment costs reduce overall expenses, but there are additional costs because of the extra feed necessary to raise the additional pigs and the costs required to house the depopulated pigs. However, it is possible that the economic benefits are from the control of other pathogens and not merely the PRRS virus. The details for nursery depopulation and cleanup protocol for the elimination of the virus are shown in Table 18-4. In an experimental infection with PRRSV, it was found that the infected pigs had greater serum concentrations of IL-1β, TNF-α, IL-12, IFN-γ, IL-10, and haptoglobin than sham controls. The results indicated that PRRSV-stimulated secretion of cytokines involved in innate, Th1, and T-reg immune responses. Mannan oligosaccharides regulated the expression of nonimmune and immune genes in pig leukocytes303 and were able to enhance the immune response without overstimulation. Mannan oligosaccharide-containing compounds were found to decrease the levels of the serum TNF-α. The levels of IL-1β and IL-12 may help to promote innate and T-cell immune functions.304 Management of the Gilt Pool Management of the gilt pool is the single most important strategy for long-term effective control. Controlling the infection in the breeding herd is a prerequisite to controlling infection in the nursery and finishing pig groups. Strategies like partial depopulation and piglet vaccination are ineffective unless the breeding herd is first stabilized, preventing piglets from becoming infected

1812

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

before weaning. Replacements are a major source of introduction of the virus and activating existing virus in the breeding herd. They also initiate the formation and maintenance of breeding herd replacements. Subpopulations are subsets of naive or recently infected gilts or sows that coexist within chronically infected herds. These subpopulations perpetuate viral transmission in the breeding herd and farrowing units, which ultimately produces successions of infected piglets before weaning. Modifications in gilt management that may minimize subpopulations include ceasing introduction of replacement animals for a 4-month period, beginning to select replacements from the finishing unit, or introducing a 4-month allotment of gilts at one time. Exposure to the virus in the breeding herd can be controlled by managing the gilt pool using two strategies. In one strategy, herds may be closed to outside replacements, and replacement males and females are raised on the farm. In the other strategy, replacement gilts are held in an off-site holding facility from 9 to 12 weeks of age until breeding age at 7 to 7.5 months, or even much earlier. This is combined with nursery depopulation as described earlier. Before entry of the gilts into the herd, they are serologically tested for evidence of seronegativity or a declining titer, which is required for entry into the herd. The gilts are isolated and quarantined for acclimatization for 45 to 60 days. This may be combined with two vaccinations, 30 days apart, after entering quarantine. This method reduces the risk of introducing potentially viremic animals into the existing population. The method selected will depend on the production system, management capabilities, and facilities available on each farm. The introduction of younger gilts, in larger groups, less frequently throughout the year, is being recognized as the most effective method for introducing replacement stock to virusinfected herds and long-term control of the disease. Controlled Infection of Breeding Herd The presence of subpopulations of highly susceptible breeding animals in the herd can be a major risk factor for maintaining viral transmission within problem herds and may explain recurrent outbreaks of reproductive failure. By intentionally exposing all members of a population to the virus, it may be possible to eliminate subpopulations and produce consistent herd immunity. In endemic herds, exposure of gilts to the virus before breeding is critical for prevention of reproductive failure. Seronegative replacement gilts can be introduced into seropositive herds at 3 to 4 months of age to allow for viral exposure before breeding. If the status is uncertain, quarantine and exposure to

nursery pigs of the importing unit is a suitable policy if replacement gilts are bought in before they are bred. It is possible to convert a PRRS-positive unit to a negative herd by managing the gilt pool and regulating the pig flow. It appears that PRRSV infection eventually either disappears or becomes inactive in the donor gilt population. Similarly, serum from nursery pigs (thought to be PRRSV viremic) given to negative replacement gilts resulted in seroconversion of all 50 gilts receiving the serum. Control of Secondary Infections When outbreaks of the disease occur in nursing piglets, and virus circulation is occurring continuously in the farrowing facility, the following are recommended: • Cross-foster piglets only during the first 24 hours of life • Prevent movement of pigs and sows between rooms • Eliminate the use of nurse sows • Euthanize piglets with low viability • Minimize injections of suckling pigs • Stop all feedback of pig and placental tissues • Follow strict all-in/all-out pig flow in the farrowing and nursery rooms. These are similar to the system developed in the United States called the McRebel system. This was a method of control showing that cross-fostering of piglets should be minimal within the first 24 hours and banned after this time. Feedback has been tried, although there are a lot of reasons not to do so. Minced whole piglets were fed to sows and the herd then closed for 23 weeks. No clinical signs were observed. One-third of the sows present at the time of the outbreak were still seropositive 20 months after the deliberate infection. Disinfection at cold temperatures was described. Biosecurity Standard methods, such as quarantining and serologic screening of imported breeding stock and restrictions on visitors, are recommended to keep units free of infection. Control of infection between herds depends on restricting the movement of pigs from infected herds to uninfected herds. If pigs have to be bought in, then seropositive animals should be imported into seropositive herds. Only seronegative boars should be allowed entry into artificial insemination units. Biosecurity practices regarding PRRSV have been investigated in Quebec in two areas of different swine density. A questionnaire was sent to 125 breeding sites and 120 growing sites. The frequency of biosecurity practices ranged from 0% to 2% for a barrier at the site entrance, 0% to 19% for showering, 20% to 25% for truck washing between loads, 51% to 57% for absence of rendering or rendering without access to the site, and 26% to

51% for absence of gilt purchase or purchase with quarantine. Better practices were found in the breeding herds. In the high-density area, there was a lower level of biosecurity on the growing sites. There were two patterns of biosecurity, a low one and a high one. For the breeding sites the higher pattern was observed when the site was away from other pig sites, more than 300 m from a public road, with a higher number of sows or being part of integrated production.305 In a second part of the study, on prevalence and risk factors, it was found that the overall prevalence of PRRS was 74.0%. Four main factors were associated with PRRS positivity, and these were large pig inventory, proximity to closest site (16%), absence of shower (27%), and free access to the site by the rendering truck (10%).305 Boar studs that are free should only import boars that are certified free from tested herds. The status of the boar stud should be tested every 2 weeks with a combination of ELISA and PCR. Testing protocols that used PCR on serum detected the PRRSV introduction earlier than the protocols that used PCR on semen, and these were earlier than those that used ELISA on serum. The most intensive protocol (testing 60 boars three times a week by PCR on serum) would need 13 days to detect 95% of the PRRSV introductions.306 A vaccination study using a modified live PRRSV vaccine on European and North American PRRSV shedding from boars showed that boar vaccination decreased the shedding of U.S. PRRSV but not the European strain.307 Vaccine and Vaccination The inefficiency of current vaccines to crossprotect against all strains of PRRSV may be caused by variability within GP5.2 Adjuvants for use in PRRSV vaccines have been reviewed.308 Of 11 adjuvants tested 5 enhanced cell-mediated immunity to PRRSV. In particular, IL-12 and CpG ODN significantly enhanced the protective efficacy of PRRSV vaccines in challenge models. The immunostimulatory oligodeoxynucleotides have been used previously.309 TLR ligands enhance the protective effects of vaccination against PRRS syndrome in swine using killed vaccines.310 Vaccination with a combined PRRSV/ MH vaccine did not differ in protective efficacy compared with the protective efficacy of the two single vaccines. This indicates that neither vaccine interfered with each other.311 Vaccine efficacy of PRRSV chimeras has been described,312 and the study suggested that only specific chimeras can attenuate clinical signs in swine and that attenuation cannot be directly linked to primary virus replication. Pigs infected with PRRSV at the time of vaccination for swine influenza had an increased level of macroscopic and microscopic pneumonia, suggesting that there was

Infectious Diseases Primarily Affecting the Reproductive System

VetBooks.ir

a reduced SIV vaccine efficiency.313 In addition, there was also increased clinical disease and shedding of SIV during the acute phase of SIV infection. Immunologic solutions for the treatment and prevention of PRRSV have been reviewed.314 No differences were found between intradermal and IM vaccinated pigs and those subsequently exposed to a heterologous Italian strain.315 The antibody response and the maternal immunity when PRRSV-immune sows were boosted with experimental farm-specific and commercial PRRSV vaccines has been described.316 The study was designed to boost PRRS-immune sows against circulating viruses. Three PRRSV isolates were taken. Booster vaccinations used either commercial vaccines or inactivated farm-specific isolate vaccines. A boost was found in all three farm-specific vaccinations. The commercial attenuated vaccine boostered immunity in 2/3 herds but the commercial nonattenuated dead vaccine did not affect the immunity on any of the three farms. In a second part of the study, similar vaccines were given at 60 days’ gestation. The farm-specific vaccines produced a significant increase in farmspecific neutralizing antibodies in all sows. Virus-neutralizing antibodies were also transferred to the piglets via colostrum and were detectable in the serum of these animals until 5 weeks after parturition. Not all sows vaccinated with the commercial attenuated vaccine showed an increase in the farmspecific virus-neutralizing antibodies, and the piglets in this group received a lower level of colostral antibodies. The number of viremic animals was significantly lower in the piglets of both groups of vaccinated animals than among mock vaccinated animals until at least 9 weeks of age. Vaccination of Gilts The two commercial modified live virus vaccines against PRRSV in pregnant gilts were shown to replicate in pregnant gilts and to cross the placenta.317 It was concluded that the vaccines had no marked detrimental effects in pregnant gilts but that they could cross the placenta and lead to the birth of congenitally affected piglets. Intranasal delivery of PRRS-MLV with a potent adjuvant (from M. tuberculosis wholecell lysate) to elicit cross-protective immunity to a heterologous strain of PRRSV generated effective cross-immunity. There was reduced lung pathology, enhanced neutralizing antibodies, and reduced viremia. There was a reduced secretion of immunosuppressive cytokines (IL-10 and TGF-β) and an upregulation of the Th-1 cytokine IFN-γ in blood and lungs.318 The ORF5a antibody response is neither neutralizing nor protective.319 Vaccination is an aid to management in developing effective immunity. The goal is to produce a constant level of immunity across

a defined population. This effectively immunizes the entire population and eliminates the nonimmune, susceptible subpopulations. Vaccination is most effective when used in replacement gilts combined with adequate isolation and acclimatization and in sows after farrowing and prebreeding. The routine vaccination of sows is not economically viable in herds affected with PRRSV. The vaccine is best suited for stabilizing the herd and is a necessity before nursery depopulation or commingling segregated early weaning piglets from virus-positive herds. Vaccination is also intended to produce protective immunity in weaned and growing pigs. The PRRS virus exists in many forms and therefore the closer the genetic makeup between the immunizing virus and the challenge virus the better. Both inactivated and modified live virus vaccines are available. Previous vaccination with a live attenuated strain produced an increase in proinflammatory cytokines and proimmune cytokine gene expression. In addition, a higher level of cortisol production suggested that there was an activation of the hypothalamus-pituitary-adrenal axis response. Vaccination produces an early immune response in pigs and a more efficient control of inflammation.320 Inactivated Vaccines Immunization of pigs with a genotype I attenuated vaccine provided partial protection against challenge with a highly virulent genotype II strain. There was a lower mortality, fewer days of fever, lower frequency of catarrhal bronchopneumonia, higher weight gains and lower viremia compared with unvaccinated control pigs.321 Killed vaccines that are inactivated using methods that preserve the PRRSV entryassociated domains are most useful for the development of effective inactivated vaccines because they facilitate internalization into macrophages.322 An experimental inactivated PRRSV vaccine that induces virusneutralizing antibodies has been described.323 The vaccine uses an optimized inactivation procedure and a suitable adjuvant, and by using these methods it was shown that inactivated PRRS vaccines can be developed that induce virus-neutralizing antibodies and offer partial protection on challenge. Killed vaccines may not produce a measurable antibody response stimulation, but activation of lymphocytes does occur and any subsequent exposure with vaccine or field virus increases that response. There is no possibility of producing a viremia and no chance of producing shedding, and there are no detrimental effects on the host. However, there is no evidence that killed vaccines protect against heterologous challenge. A killed, oil-adjuvanted vaccine based on a Spanish isolate of the virus is intended for protection against reproductive disease in

1813

gilts and sows. Initial vaccination involves 2 vaccinations, 21 days apart, with the second vaccination at least 3 weeks before breeding and with booster vaccinations recommended during subsequent lactations. Experimental challenge provides 70% protection based on pigs born alive and surviving to 7 days. An autogenous inactivated vaccine was compared with commercial vaccines against homologous and heterologous challenge.324 In this study the experimental inactivated homologous vaccines shortened the viremia on challenge, but the experimental heterologous and commercial inactivated vaccine had no or only a limited effect on the viremia. Live Vaccines A study in China325 on farms with a complex microbial ecology showed that mass vaccination with an attenuated virus vaccine can improve health status and production performance of sows and their offspring. Modified live vaccines do give a safe and efficacious protection against a wide variety of heterologous challenge strains. The vaccine virus can be transmitted from vaccinated to naive pigs and to naive herds. Vaccination of boars causes the virus to be shed, but if they have been previously exposed and then are vaccinated then there is no release of virus. The live vaccine given to finishing pigs will protect against respiratory infections. A modified live virus vaccine given once is safe for use in pregnant sows, and vaccine virus is not transmitted to susceptible contact pigs. In growing pigs vaccinated at 3 to 18 weeks of age, the vaccine elicits protective immunity within 7 days and lasts 16 weeks. Compared with controls, vaccinated animals have a reduced level of viremia, their growth rates are superior, and they have a reduced number of lung lesions. Field trials suggest that the vaccine provides protection to nursery pigs in units with endemic infection. Live viral vaccines in sows may or may not be a good idea because they demonstrated that reduced numbers of pigs were born alive and there were increased numbers of stillborn piglets to vaccinated sows irrespective of the stage of vaccination. Both single-strain and multistrain vaccines can be attenuated and be useful immunogens, but additional studies are needed to make sure that the multistrain vaccines can be recommended for routine field use. In Denmark in 1996, the use of a modified live virus vaccine licensed for use in pigs 3 to 18 weeks of age was used in a large number of PRRSV seropositive herds. Following vaccination, a large number of herds experienced an increased incidence of abortions, stillbirths, and poor performance during the nursery period. The vaccine virus was isolated from fetuses, and it was concluded that the virus was transmitted to seronegative nonvaccinated pregnant gilts and sows (see the section Methods of

1814

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

Transmission). The viruses were collected and sequenced and shown to have only a 60% homology to Lelystad virus, the European type strain, but a 98.5% homology to strain ATC-2332, which is the North American reference strain. It was therefore thought that the vaccine viruses were reverting to their natural antecedents and their virulence. Describing the vaccine virus it was shown that given to piglets it could infect nonvaccinated sows. Given to sows it can produce congenital infection, fetal death, and an increased preweaning mortality. The vaccine virus can be maintained in the population where it may undergo considerable genetic change and then lead to the establishment of new variants. Vaccination with the U.S. type vaccine produces little effect on viremia with EU PRRSV. Vaccination with EU type vaccines produced complete suppression of EU PRRSV isolates. A modified live virus vaccine has been evaluated in pigs vaccinated at 3 weeks of age and challenged at 7 weeks of age. Efficacy was evaluated using homologous and heterologous strains of virus known to cause respiratory and reproductive disease. The vaccine controlled respiratory disease but did not prevent infection and viremia. There are no published reports of randomized clinical trials evaluating the vaccines under naturally occurring conditions. In many cases of PRDC, vaccination fails simply because it was given too late or because there was no cross-protection to heterologous strains. DNA vaccination is said to produce both humoral and cellular responses and neutralization epitopes on the viral envelope glycoproteins encoded by ORF4. Possibly recombinants can be used as vaccines. In a survey in Germany, 18.5% of the samples were positive for the EU wild-type virus, EU genotype vaccine virus was detected in 1.3%, and the North American genotype vaccine virus was found in 8.9% of all samples. North American vaccine virus was frequently detected in nonvaccinated animals.326 The first modified-live vaccine was first released in 1994 and since then a number of other modified live and killed-virus vaccines have been developed. Vaccines should induce rapid immunity, have no adverse reactions, and be able to differentiate vaccinated from naturally infected animals (DIVA vaccine).308,327,328 Mass vaccination using modified live virus against homologous infection was shown to be effective in reducing economic losses from PRRSV. It did not eliminate the virus but it did reduce viral shedding 97 DPI.329 Two vaccines were compared (one inactivated and one modified live), and the modified live virus was the only type of vaccine capable of establishing protective immunity as measured by viral load in blood and tissues. The inactivated vaccine evoked no measurable protective immunity. The

modified live vaccine seemed to be based on cell-mediated immunity.330 A modified live vaccine partially protected a group of pigs given a heterologous virus vaccine; intervention reduced the duration of shedding but did not reduce the viral load in tissues or the proportion of persistently infected pigs. When the pigs were subsequently given the highly virulent virus, infection and shedding were not prevented.331 The modified live vaccines for PRRSV have been reviewed.332 None of the vaccines studied (Ingelvac PRRS MLV, Amervac PRRS, Pyrsvac-183, and Porcilis PRRS by the IM route) caused detectable clinical signs in vaccinated pigs, although lung lesions were found. Neither Pyrsivac-183 nor Porcilis PRRS could be detected in the pulmonary alveolar macrophages or in lung sections by IHC, suggesting that these viruses may have lost their ability to replicate in PAM. In these pigs, there was also a lower transmission rate and a delay in the onset of viremia, which may be explained by the lack of infection and therefore replication in the alveolar macrophage. Novel strategies for the next generation of vaccines have been described333 and stress the future importance of reverse genetics system-based vaccine development. Serologic marker candidates have been identified.334,335 Vectored vaccines may have a place in the future.336-338 Recombinant fowlpox virus-based virus with coexpression of GP5/GP3 proteins of PRRSV and swine IL-18 has been described339 as potential vaccines. The fusion of the heat shock protein (HSP70) of H. parasuis with GP3 and GP5 of PRRSV enhanced the immune responses and protective efficacy of a vaccine.340 The strategy of coexpressing GPGP-linked GP5 and M fusion protein may be a promising approach for future PRRSV vaccine development.341 A canine adenovirus has also been used as a vehicle.342 Overattenuation of an HP-PRRSV (over 100 passages) was used to produce a possible vaccine343 suggesting that loss of pathogenicity has to be balanced with loss of antigenicity. Vaccination against PRRSV resulted in significantly lower viral loads of PCV2 in animals over 13 weeks compared with nonvaccinated animals but it had no effect on quantitative PCR results for PRRSV in 4- to 12-week-old pigs. PRRS vaccinates had significantly lower levels of PCV2 viral loads when peak wasting disease was seen.344 Concurrent PRRSV and PCV2 vaccination produced no interference with the development of the specific humoral and cell-mediated immunity and is associated with clinical protection on natural challenge.345 PRRSV vaccine induced a low but significant virus-specific response IFN-γ secreting cell response on stimulation with

both the vaccine strain and two heterologous PRRSV isolates.346 An isolate of PRRSV has been shown to produce IFNs and may be useful for the development of vaccines.347 Vaccination Against High Pathogenicity Porcine Reproductive and Respiratory Syndrome A live attenuated vaccine was successfully produced from an HP-PRRSV strain TJ and the attenuation produced a further 120 amino acid deletion as well as the 30 amino acid deletion found in these HP-PRRSV strains.348 The pigs were protected from the lethal challenge and did not develop fever and clinical disease. The vaccinated pigs also gained more weight and had milder pathologic lesions. The effective protection lasted at least 4 months. A live attenuated vaccine has been used against HP-PRRSV.349 Vaccination of Boars The use of an attenuated virus vaccine in boars resulted in a marked reduction in viremia and shedding of the virus in semen compared with nonvaccinated control animals. Introducing a vaccination program using the live virus vaccine may be considered as a potential method to reduce the risk of transmission of virus by artificial insemination. In contrast, no changes in onset, level, and duration of viremia, or shedding of virus in semen, were observed using the inactivated virus vaccine. FURTHER READING Dee S, et al. Use of a production region model to assess the efficacy of various air filtration systems for preventing airborne transmission of PRRS and M. hyopneumoniae: Results from a 2 year study. Virus Res. 2010;154:177-184. Dokland T. The structural biology of PRRSV. Virus Res. 2010;154:86-97. Frossard J-P. Porcine reproductive and respiratory syndrome virus evolution and its effect on control strategies. Pig J. 2013;68:20-25. Gomez-Laguna J, et al. Immunopathogenesis of PRRSV in the respiratory tract of pigs. Vet J. 2013;195: 148. Karniychuk UU, Nauwynck HJ. Pathogenesis and prevention of placental and transplacental porcine reproductive and respiratory syndrome virus infection. Vet Res. 2013;44:95. Murtaugh MP, Genzow M. Immunological solutions for treatment and prevention of PRRS. Vaccine. 2011;29:8192-8204. Murtaugh MP, et al. The ever-expanding diversity of PRRSV. Virus Res. 2010;154:18-30. Nauwynck HJ, et al. Microdissecting the pathogenesis and immune response of PRRSV infection paves the way for more efficient PRRSV vaccines. Transboundary Emerg Dis. 2012;59(suppl 1):50-54. Sang Y, et al. Interaction between innate immunity and PRRSV. Anim Health Res Rev. 2011;12:149-167. Shi M, et al. Molecular epidemiology of PRRSV: A phylogenetic perspective. Virus Res. 2010;154:7-17. Thanawongnuwech R, Suradhat S. Taming PRRSV: Revisiting the control strategies and vaccine design. Virus Res. 2010;154:133-140.

Infectious Diseases Primarily Affecting the Reproductive System

VetBooks.ir

Yoo D, et al. Modulation of host cell responses and evasion strategies for PRRSV. Virus Res. 2010;154:48-60. Zhou L, Yang H. PRRSV in China. Virus Res. 2010;154:31-37.

REFERENCES

1. Opriessnig T, et al. Anim Health Res Rev. 2011;12:133. 2. Murtaugh MP, et al. Virus Res. 2010;154:18. 3. Gorbalenya AE, et al. Virus Res. 2006;117:17. 4. Fang Y, et al. Arch Virol. 2007;152:1009. 5. Shi M, et al. Virus Res. 2010;154:7. 6. Martinez-Lobo FJ, et al. Vaccine. 2011;29:6928. 7. Fang Y, Snijder EJ. Virus Res. 2010;154:61. 8. Firth AE, et al. J Gen Virol. 2011;92:1097. 9. Johnson CR, et al. J Gen Virol. 2011;92:1107. 10. Wei Z, et al. J Virol. 2012;86:9941. 11. Xia Pa, et al. Vet Microbiol. 2009;138:297. 12. Lee C, Yoo D. Virology. 2006;346:238. 13. Kim W-I, et al. Vet Microbiol. 2013;162:10. 14. de Lima M, et al. Virology. 2009;390:31. 15. Brockmeier S, et al. Virus Res. 2012;169:212. 16. Johnston CR, et al. J Gen Virol. 2011;92:1107. 17. Wenhui L, et al. J Virol. 2012;86:9543. 18. Stadejek T, et al. J Gen Virol. 2006;87:1835. 19. Costers S, et al. Virus Res. 2010;154:104. 20. Kvisvgaard LK, et al. Virus Res. 2013;178:197. 21. Kvivsgaard LK, et al. Vet Microbiol. 2013;167:334. 22. Zhu L, et al. Vet Microbiol. 2011;147:274. 23. Li Y, et al. Vet Microbiol. 2009;138:150. 24. Cha S-H, et al. Vet Microbiol. 2006;117:248. 25. Frossard J-P, et al. Vet Microbiol. 2012;158:308. 26. Frossard J-P, et al. Vet Microbiol. 2013;162:507. 27. Mateu E, et al. Virus Res. 2006;115:198. 28. Prieto C, et al. Vet J. 2009;180:363. 29. Carlsson U, et al. Transbound Emerg Dis. 2009;56:121. 30. Amonsin A, et al. Virol J. 2009;6:143. 31. Tian K, et al. PLoS ONE. 2007;2:3526. 32. Xiao XL, et al. J Virol Methods. 2008;149:49. 33. Normile D. Science. 2007;317:1017. 34. Feng Y, et al. Emerg Infect Dis. 2008;14:1774. 35. Wu J, et al. Arch Virol. 2009;154:1589. 36. Li B, et al. Vet Microbiol. 2010;146:226. 37. Wang L, et al. J Virol. 2012;86:13121. 38. Zhang G, et al. J Virol. 2012;86:11396. 39. Li B, et al. J Clin Microbiol. 2011;49:3175. 40. Kim HK, et al. Vet Microbiol. 2011;150:230. 41. Kim S-H, et al. Vet Microbiol. 2010;143:394. 42. Miller LC, et al. Vet Res. 2012;8:208. 43. Xiao S, et al. BMC Genomics. 2010;11:544. 44. Hu SP, et al. Transbound Emerg Dis. 2012;60:351. 45. Lv J, et al. J Gen Virol. 2008;89:2075. 46. Shen J, et al. Genome Announc. 2013;1:e00486-13. 47. Zhou L, et al. Virus Res. 2009;145:97. 48. Zhou Y-J, et al. Virus Res. 2009;144:136. 49. Yu X, et al. Vet Microbiol. 2012;158:291. 50. Metwally S, et al. Transbound Emerg Dis. 2010;57:315. 51. Guo B, et al. Virology. 2013;446:238. 52. Song T, et al. J Virol. 2012;86:4040. 53. Guo B, et al. Virology. 2013;435:372. 54. Descotes J, Gourand A. Expert Opin Drug Metab Toxicol. 2008;4:1537. 55. Tarrant JM. Toxicol Sci. 2010;117:4. 56. Sun Y, et al. Viruses. 2012;4:424. 57. Behrens EM, et al. J Clin Invest. 2011;121:2264. 58. Nieuwehhuis N, et al. Vet Rec. 2012;170:225. 59. Corbellini LG, et al. Vet Microbiol. 2006;118:267. 60. Lopez-Soria S, et al. Transbound Emerg Dis. 2010;57:171. 61. Reiner G, et al. Vet Microbiol. 2009;136:250. 62. Greiser-Wilke I, et al. Vet Microbiol. 2010;143:213. 63. Kinman TG, et al. Vaccine. 2009;8:2704. 64. Rosendal T, et al. Can J Vet Res. 2010;74:118.

65. Brar MS, et al. J Gen Virol. 2011;92:1391. 66. Evans CM, et al. Vet Res. 2008;4:48. 67. Holtkamp DJ, et al. Prev Vet Med. 2010;96:186. 68. Lambert M-E, et al. Vet Res. 2012;8:76. 69. Velasova M, et al. Vet Res. 2012;8:184. 70. Lewis CRG, et al. J Anim Sci. 2009;87:876. 71. Doeschl-Wilson AB, et al. J Anim Sci. 2009;87:1638. 72. Badaoui B, et al. BMC Vet Res. 2013;9:58. 73. Kwong GPS, et al. Prev Vet Med. 2013;110:405. 74. Charpin C, et al. Vet Res. 2012;43:69. 75. Evans CM, et al. Prev Vet Med. 2010;93:248. 76. Dee S, et al. Can J Vet Res. 2006;69:64. 77. Pitkin A, et al. Vet Microbiol. 2009;136:1. 78. Hermann J, et al. Vet Res. 2007;38:81. 79. Otake S, et al. Vet Microbiol. 2010;145:198. 80. Pitkin A, et al. Vet Microbiol. 2009;136:1. 81. Cho JG, et al. Can J Vet Res. 2006;70:297. 82. Pitkin A, et al. Vet Microbiol. 2009;136:1. 83. Cutler TD, et al. Vet Microbiol. 2011;151:229. 84. Dee S, et al. Vet Res. 2009;40:39. 85. Pitkin A, et al. Can J Vet Res. 2009;73:298. 86. Cano JP, et al. Vet Rec. 2007;160:907. 87. Molina RM, et al. Transbound Emerg Dis. 2008;56:1. 88. Pitkin A, et al. Can J Vet Res. 2009;73:91. 89. De Baere MI, et al. Vet Res. 2012;43:47. 90. Wang X, et al. Arch Virol. 2007;152:289. 91. Gomez-Laguna J, et al. Transbound Emerg Dis. 2012;10:1865. 92. Klionsky DJ. Nat Rev Mol Cell Biol. 2007;8:931. 93. Liu Q, et al. Virology. 2012;429:136. 94. Butler JE, et al. J Immunol. 2007;178:6320. 95. An T-Q, et al. Vet Microbiol. 2010;143:371. 96. Kim JK, et al. J Virol. 2006;80:689. 97. De Baere MI, et al. Vet Res. 2012;43:47. 98. Welch S-K, Calvert JG. Virus Res. 2010;154:98. 99. Calvert JG, et al. J Virol. 2007;81:7371. 100. Cafruny WA, et al. Virol J. 2007;3:90. 101. Lee YJ, Lee C. Vet Immunol Immunopathol. 2012;150:213. 102. Patton JB, et al. Virus Res. 2009;140:161. 103. Gruenberg J, van der Goot FG. Nat Rev Mol Cell Biol. 2006;7:495. 104. Misinzo GM, et al. Vet Res. 2008;39:55. 105. Van Gortp H, et al. Arch Virol. 2009;154:1939. 106. Van Breedam W, et al. J Gen Virol. 2010;91:1659. 107. Music N, Gagnon CA. Anim Health Res Rev. 2010;11:135. 108. Lee C, et al. Virology. 2006;346:238. 109. Pei Y, et al. Virus Res. 2008;135:107. 110. Kwon B, et al. Virology. 2008;380:371. 111. Wang C, et al. Vet Microbiol. 2008;131:339. 112. Faaberg KS, et al. Virus Res. 2010;154:77. 113. Rowland RR. Virus Res. 2010;154:1. 114. Zhang X, Moser DM. J Pathol. 2008;214:161. 115. Lee YJ, Lee C. Virus Res. 2010;152:50. 116. Lee C, Yoo D. Virology. 2006;355:30. 117. Du Y, et al. Virus Res. 2010;147:294. 118. Chang HC, et al. Vet Microbiol. 2008;129:281. 119. Flores-Mendoza L, et al. Clin Vaccine Immunol. 2008;15:720. 120. Loving CI, et al. Immunology. 2007;120:217. 121. Gimeno M, et al. Vet Res. 2011;42:9. 122. Gomez-Laguna J, et al. J Comp Pathol. 2010;142:51. 123. Zhang Y, et al. Vet Microbiol. 2012;160:473. 124. Sagong M, Lee C. Virus Res. 2010;151:88. 125. Li Y, et al. J Gen Virol. 2012;93:829. 126. Chaung H-C, et al. Comp Immunol Microbiol Infect Dis. 2010;33:197. 127. Liu C-H, et al. Vet Microbiol. 2009;136:266. 128. Manickam C, et al. Vet Microbiol. 2013;162:68. 129. Silva-Campo E, et al. Virology. 2012;430:73. 130. Pintaric M, et al. Vet Immunol Immunopathol. 2008;121:61.

1815

131. Lodoen MB, Lainier LL. Curr Opin Immunol. 2006;18:391. 132. Costers S, et al. Arch Virol. 2008;153:1453. 133. Cao J, et al. Vet Microbiol. 2013;164:261. 134. Caliguri MA. Blood. 2008;112:461. 135. Renukaradhya GJ, et al. Viral Immunol. 2010;23:457. 136. Dvivedi V, et al. Virol J. 2012;9:45. 137. Cecere TE, et al. Vet Microbiol. 2012;160:233. 138. Silva-Campo E, et al. Virology. 2009;387:373. 139. Silva-Campo E, et al. Virology. 2010;396:264. 140. Silva-Campo E, et al. Virology. 2012;85:23. 141. Rodriguez-Gomez IM, et al. Transbound Emerg Dis. 2013;60:425. 142. Halstead SB, et al. Lancet Infect Dis. 2010;10:712. 143. Belkaid Y. Nat Rev Immunol. 2007;7:875. 144. Dwivedi V, et al. Vaccine. 2011;29:4067. 145. LeRoith T, et al. Vet Immunol Immunopathol. 2011;140:312. 146. Wongyanin P, et al. Vet Immunol Immunopathol. 2010;132:170. 147. Silva-Campo E, et al. Virology. 2012;430:73. 148. Chen Z, et al. J Gen Virol. 2010;91:1047. 149. Costers S, et al. Vet Res. 2009;40:46. 150. Gomez-Laguna J, et al. Comp Immunol Microbiol Infect Dis. 2011;34:143. 151. Subramaniam S, et al. Virology. 2010;406:270. 152. Subramaniam S, et al. Virology. 2012;432:241. 153. Barranco I, et al. Vet Immunol Immunopathol. 2012;149:262. 154. Gomez-Laguna J. Vet Microbiol. 2012;158:187. 155. Backus GS, et al. Environ Health Perspect. 2010;118:1721. 156. Diaz I, et al. Vet Res. 2012;43:30. 157. Weaver LK, et al. J Leucocyte Biol. 2007;81:663. 158. Hou J, et al. Virol J. 2012;9:165. 159. Gomez-Laguna J, et al. Vet J. 2013;195:148. 160. Lunney JK, et al. Virus Res. 2010;154:185. 161. Beura LK, et al. J Virol. 2010;84:1574. 162. Subramaniam S, et al. Proc 90th Meet Conf Res Work Anim Dis, Chicago 2009; Abstr. 176. 163. Overend C, et al. J Gen Virol. 2007;88:925. 164. Bao D, et al. Vet Immunol Immunopathol. 2013;156:128. 165. Shi X, et al. Virus Res. 2010;153:151. 166. Calzada-Nova G, et al. Vet Immunol Immunopathol. 2010;135:20. 167. Peng Y-T, et al. Vet Microbiol. 2009;136:359. 168. Miguel JC, et al. Vet Immunol Immunopathol. 2010;135:314. 169. Klinge KL, et al. Virol J. 2009;6:177. 170. Dotti S, et al. Res Vet Sci. 2013;94:510. 171. Beura LK, et al. J Virol. 2011;85:12939. 172. Beura LK, et al. J Virol. 2010;84:1574. 173. Bowie AG, Unterholzer L. Nat Rev Immunol. 2008;8:911. 174. Kawai T, Akira S. Int Immunol. 2009;21:317. 175. Beura LK, et al. Virology. 2012;433:431. 176. Versteeg GA, GarciaSastre A. Curr Opin Microbiol. 2010;13:508. 177. Beura LK, et al. J Virol. 2010;84:1574. 178. Song C, et al. Virology. 2010;407:268. 179. Patel D, et al. J Virol. 2010;84:11405. 180. Li H, et al. J Gen Virol. 2010;81:2947. 181. Sun Z, et al. J Virol. 2010;84:7832. 182. Kimman TG, et al. Vaccine. 2009;27:315. 183. Nan Y, et al. Virology. 2012;43:261. 184. Yoo D, et al. Virus Res. 2010;154:48. 185. Zhou Y, et al. Can J Vet Res. 2012;76:255. 186. Chen Z, et al. Virology. 2010;198:87. 187. Huber JP, Farrar JD. Immunology. 2011;132:446. 188. Luo R, et al. Mol Immunol. 2008;45:2839. 189. Brockmeier SL, et al. Viral Immunol. 2009;22:173. 190. Brockmeier SL, et al. Clin Vaccine Immunol. 2012;19:508. 191. Kim O, et al. Virology. 2010;402:315.

1816

VetBooks.ir

192. 193. 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211. 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. 224. 225. 226. 227. 228. 229. 230. 231. 232. 233. 234. 235. 236. 237. 238. 239. 240. 241. 242. 243. 244. 245. 246. 247. 248. 249. 250. 251. 252. 253. 254. 255. 256. 257. 258.

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

Sagong M, Lee C. Arch Virol. 2011;156:2187. Lee YJ, Lee C. Virology. 2012;427:80. Ait-Ali T, et al. Immunogenetics. 2011;63:437. Sun Z, et al. J Virol. 2012;86:3839. Barranco I, et al. Transbound Emerg Dis. 2012;59:145. Van Gorp H, et al. J Gen Virol. 2008;89:2943. Dong S, et al. Res Vet Sci. 2012;93:1060. Gudmundsdottir I, Risatti GR. Virus Res. 2009;145:145. Hou J, et al. Virus Res. 2012;167:106. Gomez-Laguna J, et al. Comp Immunol Microbiol Infect Dis. 2010;33:51. Gomez-Laguna J, et al. Vet Microbiol. 2012;158:187. Darwich L, et al. Vet Microbiol. 2011;150:49. Mateu E, Diaz I. Vet J. 2008;177:345. Dotti S, et al. Res Vet Sci. 2011;90:218. Martinez-Lobo FJ, et al. Vet Microbiol. 2011;154:58. Garcia-Nicolas O, et al. Virus Res. 2014;179:204. Weesendorp E, et al. Vet Microbiol. 2013;163:1. Weesendorp E, et al. Vet Microbiol. 2013;167:638. Kim W-I, et al. Vet Micrbiol. 2007;123:10. Frydas IS, et al. Vet Res. 2013;44:73. Wang G, et al. Virol J. 2014;11:2. Wang G, et al. Vet Immunol Immunopathol. 2011;142:170. Han K, et al. J Comp Pathol. 2012;147:275. Hou J, et al. Virus Res. 2012;167:10. Li L, et al. Virol J. 2012;9:203. Cao J, et al. J Vet Diagn Invest. 2012;24:767. Guo B, et al. Virology. 2013;435:372. Mege JL, et al. Lancet Infect Dis. 2006;6:557. Borghetti P, et al. Comp Immunol Microbiol Infect Dis. 2011;34:143. Parcina M, et al. J Immunol. 2013;190:1591. Butler JE, et al. J Immunol. 2007;178:6329. Butler JE, et al. J Immunol. 2008;180:2347. Sun XZ, et al. Vaccine. 2012;30:3646. Parida R, et al. Virus Res. 2012;169:13. Diaz I, et al. Virus Res. 2010;154:61. Han J, et al. J Virol. 2010;84:10102. Kinman TG, et al. Vaccine. 2009;27:3704. Li J, Murtaugh MP. Virology. 2012;433:367. Tong GZ, et al. Emerg Infect Dis. 2007;13:1434. An T-Q, et al. Emerg Infect Dis. 2010;16:365. Li L, et al. Virus Res. 2010;154. Murtaugh M Proc 40th Ann Meet Am Assoc Swine Vet, Dallas, TX, 459. Qiao S, et al. Vet Microbiol. 2011;149:213. Diaz I, et al. Vet Res. 2012;43:30. Jung K, et al. J Gen Virol. 2009;90:2713. Wagner J, et al. Vet J. 2011;187:310. Beilage EG, et al. Tierartzl Prax. 2007;35:294. Cano JP, et al. Can J Vet Res. 2009;73:303. Lewis CRG, et al. Anim Prod Sci. 2010;50:890. Han K, et al. J Comp Pathol. 2013;148:396. Karniychuk U, et al. Microb Pathog. 2011;51:194. Karniychuk U, Nauwynck HJ. Vet Res. 2013;44:95. Kang I, et al. Res Vet Sci. 2010;88:304. Han K, et al. Clin Vaccine Immunol. 2012;19:319. Burgara-Estrella A, et al. Transbound Emerg Dis. 2012;59:532. Schulze M, et al. Acta Vet Scand. 2013;55:16. Stadejek T, et al. Vet Rec. 2011;169:441. Xiao S, et al. BMC Genomics. 2010;11:544. Hu SP, et al. Transbound Emerg Dis. 2012;10:1865. Lawson S, et al. Vaccine. 2010;28:5356. Lee YJ, et al. J Virol Methods. 2010;163:410. Wernike K, et al. J Vet Diagn Invest. 2012;24:855. Rovira A, et al. J Vet Diagn Invest. 2007;19:502. Gerber PF, et al. J Clin Microbiol. 2013;51:547. Gerber PF, et al. J Virol Methods. 2014;197:63. Li Q, et al. J Virol Methods. 2009;155:55. Rovira A, et al. J Vet Diagn Invest. 2009;21:350.

259. Lurchachaiwong W, et al. Lett Appl Microbiol. 2008;46:55. 260. Balka G, et al. J Virol Methods. 2009;115:1. 261. Chen N-H, et al. J Virol Methods. 2009;161:192. 262. Ren X, et al. J Clin Microbiol. 2010;48:1875. 263. Van Breedam W, et al. Vet Immunol Immunopathol. 2011;141:246. 264. Diaz I, et al. J Vet Diagn Invest. 2012;24:344. 265. Kittawornrat A, et al. Vet Res. 2013;9:61. 266. Okinaga T, et al. Vet Rec. 2009;164:455. 267. Lin K, et al. J Clin Microbiol. 2011;49:3184. 268. Gutierrez AM, et al. Vet Immunol Immunopathol. 2009;132:218. 269. Gomez-Laguna J, et al. Vet J. 2010;85:83. 270. Kittawornrat A, et al. Vet Microbiol. 2014;168:331. 271. Chittick WA, et al. J Vet Diagn Invest. 2011;23:248. 272. Prickett JR, et al. J Swine Health Prod. 2008;16:86. 273. Prickett JR, et al. J Vet Diagn Invest. 2008;20:156. 274. Prickett JR, Zimmerman JJ. Anim Health Res Rev. 2010;10:1. 275. Ramirez A, et al. Prev Vet Med. 2012;104:292. 276. Kittawornrat A, et al. Virus Res. 2010;154:1700. 277. Ouyang K, et al. Clin Vaccine Immunol. 2013;20:1305. 278. Kittawornrat A, et al. J Vet Diagn Invest. 2012;24:262. 279. Olsen C, et al. J Vet Diagn Invest. 2013;25:328. 280. Kittawornrat A, et al. J Vet Diagn Invest. 2012;24:1057. 281. Costers S, et al. Vet Res. 2009;40:46. 282. Han K, et al. Vet J. 2013;195:313. 283. Gomez-Laguna J, et al. Transbound Emerg Dis. 2013;60:273. 284. Morgan SB, et al. Vet Microbiol. 2013;163:13. 285. Lee S-M, Kleiboeker SB. Virology. 2007;365:419. 286. He Y, et al. Vet Microbiol. 2012;160:455. 287. Grau-Roma L, Segales J. Vet Microbiol. 2007;119:144. 288. Han K, et al. J Vet Diagn Invest. 2012;24:719. 289. Stuart AD, et al. Pig J. 2008;61:42. 290. Jiang Y, et al. Vet Res Commun. 2010;34:607. 291. Rowland RR. Sci Direct. 2007;174:451. 292. Rowland RR, Morrison RB. Transbound Emerg Dis. 2012;59(suppl 1):55. 293. Mondaca-Fernandez E, Morrison RB. Vet Rec. 2007;161:137. 294. Wayne SR, et al. J Am Vet Med Assoc. 2012;240:876. 295. Liu Y, et al. J Anim Sci. 2013;91:5668. 296. Pitkin A, et al. Vet Microbiol. 2009;136:1. 297. Dee S, et al. Vet Microbiol. 2009;138:106. 298. Dee S, et al. Vet Rec. 2010;167:976. 299. Spronk G, et al. Vet Rec. 2010;166:758. 300. Alonso C, et al. Vet Microbiol. 2012;157:304. 301. Alonso C, et al. Prev Vet Med. 2013;112:109. 302. Alonso C, et al. Prev Vet Med. 2013;111:268. 303. Che TM, et al. J Anim Sci. 2011;89:3016. 304. Che TM, et al. J Anim Sci. 2012;90:2784. 305. Lambert M-E, et al. Prev Vet Med. 2012;104:74. 306. Rovira A, et al. J Vet Diagn Invest. 2007;19:492. 307. Han K, et al. Clin Vaccine Immunol. 2011;18:1600. 308. Charerntantanakul W. Vet Immunol Immunopathol. 2009;129:1. 309. Linghua Z, et al. Vaccine. 2007;25:1735. 310. Zhang L, et al. Vet Microbiol. 2013;164:253. 311. Drexler CS, et al. Vet Rec. 2010;166:70. 312. Thanawongnuwech R, Suradhat S. Virus Res. 2010;154:133. 313. Kitikoon P, et al. Vet Microbiol. 2009;139:235. 314. Murtaugh MP, Genzow M. Vaccine. 2011;29:8192. 315. Martelli P, et al. Vaccine. 2007;25:3400. 316. Geldhof MF, et al. Vet Microbiol. 2013;167:260. 317. Scortti M, et al. Vet J. 2006;172:506. 318. Dwivedi V, et al. Vaccine. 2011;29:4058. 319. Robinson SR, et al. Vet Microbiol. 2013;164:281.

320. Borghetti P, et al. Comp Immunol Microbiol Inf Dis. 2011;34:143. 321. Roca M, et al. Vet J. 2012;193:92. 322. Delrue I, et al. Vet Res. 2009;40:62. 323. Vanhee M, et al. Vet Res. 2009;40:63. 324. Geldof MF, et al. BMC Vet Res. 2012;8:182. 325. Zhao Z, et al. Vet Microbiol. 2012;155:247. 326. Beilage EG, et al. Prev Vet Med. 2009;92:31. 327. de Lima J, et al. Vaccine. 2008;26:3594. 328. Fang Y, et al. J Gen Virol. 2008;89:3086. 329. Cano JP, et al. Am J Vet Res. 2007;68:565. 330. Zuckerman FA, et al. Vet Microbiol. 2007;123:69. 331. Cano JP, et al. Vaccine. 2007;25:4382. 332. Martinez-Lobo F, et al. Vet Res. 2013;44:115. 333. Huang YW, Meng XJ. Virus Res. 2010;154:141. 334. de Lima M, et al. Virology. 2006;353:410. 335. Vu HLX, et al. Vaccine. 2013;31:4330. 336. Cruz JLG, et al. Virus Res. 2010;154:150. 337. Pei Y, et al. Virology. 2009;389:91. 338. Huang Q, et al. J Virol Methods. 2009;160:22. 339. Guoshan S, et al. Vaccine. 2007;25:4193. 340. Li J, et al. Vaccine. 2009;27:825. 341. Chia M-Y, et al. Vet Microbiol. 2010;146:189. 342. Cai J, et al. J Vet Med Sci. 2010;72:1035. 343. Yu X, et al. Clin Vaccine Immunol. 2013;20:613. 344. Genzow M, et al. Can J Vet Res. 2009;73:87. 345. Martelli P, et al. Vet Microbiol. 2013;162:358. 346. Ferrari L, et al. Vet Immunol Immunopathol. 2013;151:193. 347. Nan Y, et al. Virology. 2012;432:261. 348. Leng X, et al. Clin Vaccine Immunol. 2012;19:1199. 349. Tian Z-J, et al. Vet Microbiol. 2009;138:34. 350. Hu SP, et al. Transbound Emerg Dis. 2013;60:351.

MENANGLE This causative virus was first identified in a three-farm disease outbreak in New South Wales in 1997. It causes reproductive problems in pigs and congenital defects and has the fruit bat as an asymptomatic reservoir. It can cause a flu-like disease in man. Only one outbreak has been described. It normally lives asymptomatically in fruit bats.

ETIOLOGY

The causative agent is an RNA virus in the family Paramyxoviridae in the genus Rubulavirus. It is closely related to Tioman virus found in fruit bats on Tioman Island, Malaysia.

EPIDEMIOLOGY

A variety of fruit bats are seropositive, including the gray-headed flying fox, black fruit bat, and spectacled fruit bat, but the virus has not been isolated from them. These fruit bats have been found in other areas of Australia as well as the original area around Menangle, New South Wales. Bat feces and urine are probably the source of infection. Transmission from pig to pig is slow and probably requires close contact. In one building, it took a long time for the sows to become affected. It probably spreads from farm to farm via infected animals. There is no sign of persistent infection and no evidence of long-term virus shedding. Present evidence suggests that

Infectious Diseases Primarily Affecting the Reproductive System

VetBooks.ir

virus survival in the environment is short because sentinel pigs placed in an uncleaned area did not seroconvert.

CLINICAL SIGNS

There is no knowledge of the incubation period as yet. In the initial outbreak, clinical signs were seen only on the farrow-to-finish farm but infected pigs were found in all three farms. The disease was an outbreak of reproductive disease with fetal death; fetal abnormalities including congenital defects, such as skeletal and neurologic defects1; mummified fetuses; stillborn fetuses; smaller litters with fewer live piglets; and a reduced farrowing rate. The farrowing rate fell from over 80% to around a low of 38% reaching an average of 60%. Many sows returned to estrus 28 days after mating, which suggests that there has been an early death of the litter. Some sows remain in pseudopregnancy for more than 60 days. It probably crosses the placenta and spreads fetus to fetus. Once the infection became endemic in the farrow-to-finish herd the reproductive failures ceased.

PATHOLOGY

The mummified fetuses vary in size and are 30 days or older. The virus causes the degeneration of brain and spinal cord. In particular, the cerebral hemispheres and cerebellum are smaller. Occasionally there may be effusions and pulmonary hypoplasia. Eosinophilic inclusions are found in the neurons of the cerebrum and spinal cord. Sometimes there is a nonsuppurative meningitis, myocarditis, and hepatitis. Experimental infections show shedding 2 to 3 days after infection in nasal and oral secretions. A tropism for secondary lymphoid tissues and intestinal epithelium has been demonstrated.2 No lesions have been seen in piglets born alive or other postnatal pigs.

DIAGNOSIS

The diagnosis is suspected when the reproductive parameters change very suddenly, as shown earlier. Diagnosis is confirmed by virus culture, and electron microscopy and virus neutralisation tests confirm the identity of the virus. Serologic tests include ELISAs, and the best way to test the herd is to use this for the sows for antibody.

DIFFERENTIAL DIAGNOSIS

The differential diagnosis includes porcine parvo virus (PPV), classical swine fever (CSF), porcine reproductive and respiratory syndrome (PRRS), encephalomyocarditis virus (EMCV), pseudorabies virus (PRV), Japanese encephalitis, swine influenza virus (SIV), and blue eye. Noninfectious causes such as toxins or nutritional deficiencies should also be considered.

1817

TREATMENT

DIAGNOSIS

It seems likely that young pigs are infected by the virus when the maternal antibody concentration declines at 14 to 16 weeks of age. By the time they enter the breeding herd their immunity is quite strong.

RT-PCR and nested RT-PCR can be used to detect the virus when virus isolation is negative. Antibody can be detected by haemagglutination inhibiton, ELISAs (IgM capture ELISA), and latex agglutination tests.

CONTROL

The best advice is to avoid contact with all fruit bats. FURTHER READING Philbey AW, et al. An apparently new virus (family Paramyxoviridae) infection for pigs, humans and fruit bats. Emerg Infect Dis. 1998;4:269.

REFERENCES

1. Philbey AW, et al. Aust Vet J. 2007;85:134. 2. Bowden TR, et al. J Gen Virol. 2012;93:1007.

JAPANESE ENCEPHALITIS (JE; JAPANESE B ENCEPHALITIS) Japanese encephalitis is an infectious disease primarily affecting horses and to a lesser extent pigs, with important zoonotic potential. It causes in excess of 50,000 human cases a year, with a case mortality rate of 25%. The condition in equides is associated with encephalitis and is covered in detail in Chapter 14 under Japanese encephalitis. In pigs the condition is associated with reproductive failure, whch is covered hereunder.

ETIOLOGY

The causative agent is the Japanese encephalitis virus of the family Flavivirdae, genus flavivirus. Based on the phylogenetic analysis of the viral envelope “E” gene, 5 different genotypes have been identified.

EPIDEMIOLOGY

The natural distribution range of the virus is southeast Asia and Australasia. The vectors are Culex spp and in particular C. tritaeniorhynchus. The virus activity is naturally maintained through bird–mosquito cycles with the heron family in particular. The night herons, little egrets and plumed egrets are particularly active as a reservoir. Pigs are important “amplifying hosts.” Pigs and these birds may allow the overwintering of the virus when mosquitoes are absent.

CONTROL

Live attenuated vaccines should be given to breeding stock 2 to 3 weeks before the start of the mosquito season. Attenuated and adjuvanted vaccines are also available. FURTHER READING Mackenzie JS, Williams DT. The zoonotic flaviviruses of southern, southeastern and eastern Asia and Australasia: The potential for emergent viruses. Zoonoses Public Health. 2009;56:338.

NEOSPOROSIS SYNOPSIS Etiology The protozoan parasite Neospora caninum; the dog is identified as the definitive host of N. caninum, but the main route of infection in cattle appears to be by vertical transmission. Epidemiology An infection of cattle worldwide and associated with epidemic and endemic abortion. Point source and congenital infections occur. Clinical findings Abortion in cows and perinatal mortality and encephalomyelitis in congenitally infected calves. Clinical pathology Serologic testing of maternal serum and fetal fluids. Necropsy findings Fetal lesions of multifocal nonsuppurative encephalitis, myocarditis, and/or periportal hepatitis. Infection confirmed by immunohistochemistry or polymerase chain reaction-based tools. Diagnostic confirmation A presumptive diagnosis can be based on the fetal histologic lesions and seropositivity of the dam, but the definitive diagnosis requires the demonstration of the parasite in fetal tissues by immunohistochemical labeling, coupled with serologic examinations. Control Feed hygiene and calving hygiene. Cull congenitally infected cattle.

PATHOGENESIS

Viremia results from the mosquito bite and usually nothing is seen. Occasionally there may be a mild fever. but quite often the virus goes straight to the testicles and causes an orchitis.

CLINICIAL SIGNS

Fetal death is common with mummified fetuses as well as stillborn and weak pigs. Boars undergo reproductive failure.

PATHOLOGY

Largely related to the abnormal fetuses.

ETIOLOGY

Neospora caninum is a cyst-forming coccidial (apicomplexan) parasite with an indirect life cycle.1-9 N. caninum primarily infects dogs and cattle; however, it has a wide host range and infects all major domestic livestock species as well as companion animals and some wildlife animals. Dogs are the definitive host and cattle the major intermediate host. Natural infection is infrequently reported in sheep, goats, and deer.1-3 N. caninum is a sporadic cause of

1818

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

encephalomyelitis and myocarditis in several species, but its principal importance is its association with endemic and epidemic abortion in cattle. It is now the most common diagnosis for abortion in cattle in most countries.

EPIDEMIOLOGY Occurrence N. caninum was initially associated with abortion in the early 1990s in pastured cattle in Australia and New Zealand and as a major cause of abortion in dairies in the United States. Since then, abortion associated with N. caninum has been reported in many countries in cattle under varying management conditions and has a worldwide occurrence.2,3 Abortion may be epizootic or sporadic. In epizootic abortion, the number of cows aborting varies. It is usually between 5% and 10%, but up to 45% of cows may abort within a short period. The period of abortion may be a few weeks to a few months. There is no major seasonal occurrence, and abortion occurs in both beef and dairy cows. Sporadic abortions occur mainly in cows that have been infected congenitally, and seropositive cows have greater risk for repeat abortions. Seropositivity in herds can be high but varies considerably. Seropositive dams have a 3- to 7-fold greater risk of abortion than seronegative dams. Methods of Transmission There are two routes of infection of cattle. The dog is the definitive host of N. caninum. Infection of cattle can occur via the ingestion of oocytes from dog feces contaminating feed or water. However, vertical (i.e., congenital) transmission occurs in both cattle and dogs, and vertical transmission appears the major route for infection in most cattle.1-3 Live-born calves from congenitally infected cows are themselves congenitally infected; the infection is thought to be persistent and lifelong. A study conducted on two dairies found 81% of seropositive cows gave birth to congenitally infected calves.1 Seroprevalence did not increase with cow age and was stable through the study period. The probability of a calf being congenitally infected was not associated with dam age, dam lactation number, dam history of abortion, calf gender, or length of gestation. Other studies have shown that this route of transmission is highly efficient, resulting in infection of 50% to 95% of the progeny of seropositive dams. Congenital infection can result in abortion or the birth of a “normal,” infected calf, and an infected cow can give birth to a clinically normal, infected calf at one pregnancy and abort in the subsequent pregnancy.2,3 The occurrence of infection in some herds can be associated with specific family lines. Although vertical transmission is the major route of infection that leads to

sporadic abortions in cattle associated with N. caninum, epidemiologic evidence suggests that postnatal (point) infection is often the cause of outbreaks of abortion. Where dog feces are the source of infection, many cattle are often exposed, and this point source of infection commonly results in outbreaks of abortion. Farm dogs have been shown to have a higher seroprevalence to N. caninum than urban dogs, suggesting that neosporosis cycles between cattle and dogs in rural environments.4 The importance of postnatal infection versus vertical infection in the genesis of abortion may vary among countries, and be associated with differences in farm management systems.4

stillbirth and increased risk of retained placenta. Losses associated with epidemic abortion have been estimated at tens (20–85) of millions of dollars to the dairy or beef industries in Australasia and the United States. Although seropositive heifers have been reported to produce less milk than sero­ negative herdmates, this difference in milk production between seropositive and seronegative animals is not necessarily apparent in herds unaffected by an abortion problem. Study of beef cattle has suggested that seropositivity might be associated with reduction in average daily weight gain, but production performance and carcass measures are not consistently reported to be affected.

Experimental Studies Abortion has been produced by experimental challenge of fetuses and pregnant cattle with culture-derived tachyzoites of N. caninum.1 Fetal death and resorption or abortion has been reproduced in ewes challenged at 45, 65, and 90 days’ gestation, but not 120 days, and lesions resemble those of ovine toxoplasmosis.2 The disease has also been reproduced experimentally in goats,1 but the importance and prevalence of this infection in naturally occurring abortions in small ruminants remains to be determined. Contaminated placenta, milk, and colostrum can result in infection of calves less than 1 week of age.

N. caninum has a predilection for fetal chorionic epithelium and fetal placental blood vessels, producing a fetal vasculitis and inflammation and degeneration of the chorioallantois, and widespread necrosis in the placentome.6 Tachyzoites penetrate host cells and are located in a parasitophorous vacuole. They can be found in macrophages, monocytes, vascular endothelial cells, fibroblasts, hepatocytes, renal tubular cells, and in the brain of infected animals. With neuromuscular disease, cranial and spinal neural cells are infected. Cell death is caused by the replication of tachyzoites (during endodyogeny).

Risk Factors Outbreaks of abortion often appear to be point source infections, but the risk factors, other than probable mass exposure to dog feces containing sporulated N. caninum oocysts, are not known. Neosporosis in dairy herds often occurs as an epizootic, with multiple abortions occurring in a 1- to 2-month period. Severely autolytic fetuses are aborted between 5 and 7 months of pregnancy in most reports, but earlier or later abortions can occur (range is between 3 and 8.5 months of pregnancy). Endemic abortion is more likely associated with the presence of congenitally infected cattle in the herd, which are at high risk of aborting, particularly in the initial pregnancy and in the pregnancy during the first lactation.2,3 Cows that have aborted have a higher risk for abortion in subsequent pregnancies, but this risk decreases with each subsequent pregnancy. It has been postulated that immunosuppression resulting from concurrent infection with other agents, such as bovine viral diarrhea virus (BVD) , may increase the risk for infection with N. caninum and precipitate abortion outbreaks. Economic Importance Economic losses relate to abortion and costs associated with establishing the diagnosis and rebreeding or replacement costs.5 Seropositivity is also associated with increased risk of

PATHOGENESIS

CLINICAL FINDINGS

Abortion is the cardinal clinical sign observed in infected cows.2,3 Fetuses may die in utero, or can be reabsorbed, mummified, stillborn, born alive but diseased, or born clinically normal but infected. Cows that are infected can have decreased milk production in the first lactation, producing approximately 1 L less of milk per cow per day than uninfected cows, are prone to abort, and have a higher risk of being culled from the herd at an early age. In addition to the occurrence of early abortion, the disease in beef herds is associated with the birth of live-born, premature, low birthweight calves. Depending on the degree of prematurity, these calves can be kept alive with intensive care during the neonatal period. Most congenitally infected calves are born alive without clinical signs. Occasionally, congenital infection can be manifest with ataxia, loss of conscious proprioception, paralysis, and/or other neurologic deficits in new-born calves,2 but most congenitally infected calves appear as clinically normal and, surprisingly, some evidence suggests that congenital infection does not necessarily have a detrimental effect on calf health and survival.3 N. caninum infection has been demonstrated in the nervous system of a horse with progressive debilitation, followed by a sudden onset of neurologic disease with paraplegia. It appears to be a rare cause of

Infectious Diseases Primarily Affecting the Reproductive System

VetBooks.ir

neurologic disease in horses, but should be considered in the differential diagnosis of equine protozoal myeloencephalitis.

CONTROL

Gross findings are not specific and the fetus may be fresh, autolyzed, or in early stages of mummification; in the placenta, the cotyledons are usually necrotic.10 The brain may be autolyzed, but should still be submitted for examination as well as the heart, liver, and placenta, if available. Histologic findings commonly relate to multifocal nonsuppurative encephalitis, myocarditis hepatitis, and/or placentitis Liver lesions may be more prominent in epizootic abortions. IHC or PCR can be used to detect tachyzoites or their DNA in tissues (particularly in the brain).7 IHC can be specific, but insensitive for identifying Neospora in the placenta; therefore, maternal serology should be used in conjunction.

All efforts should be made to exclude the possibility of dog fecal contamination of cattle feed and water and of the grazing environment.4 Placentas, aborted fetuses, and dead calves should be removed immediately and disposed of so that the definitive host and cattle cannot gain access to them. Congenitally infected cows are at high risk of abortion, and abortion rates in infected herds can be substantially reduced by culling infected animals.2-4 Congenitally infected calves can be identified by testing precolostral blood samples using a specific and sensitive serologic test and culled at a young age. If precolostral blood sampling is not feasible, examination of sera at 6 months of age should identify infected calves, with positive titers indicating either congenital infection or postnatal infection. Calves introduced into a herd should be seronegative. It is possible that strategic therapy of pregnant cows with an appropriate antiprotozoal drug could abort the infection. This could be effective in beef cattle, but would probably not be legal or appropriate in lactating dairy cattle. Although evidence for increased risk for Neospora abortion caused by immunosuppression resulting from concurrent infection with BVD virus is equivocal, control of BVD infections should be a component of antineosporosis control. There has been a considerable effort to develop vaccines against neosporosis.8,9 An inactivated tachyzoite vaccine was approved in the United States for use in pregnant cows. There are no controlled studies on its efficacy in mitigating the effects of bovine neosporosis in dairy cattle. Vaccination of dairy cattle may interfere with a herd test and cull policy.

TREATMENT

FURTHER READING

CLINICAL PATHOLOGY

Serologic testing can be conducted using IFAT or ELISA, and there appears to be good agreement in results between the two tests. ELISA using recombinant protein appears to have a higher diagnostic specificity and sensitivity than using whole-tachyzoite lysates.7 IFAT is commonly used and achieves a relatively high diagnostic specificity and sensitivity for the detection of maternal infection.7 The persistence of serum antibody titers following infection is uncertain, and they might fluctuate during pregnancy. A positive titer in a cow that has aborted indicates exposure but not causality. IgG avidity patterns have been used to predict the duration of infection. Diagnosis can also be conducted by detecting anti-N. caninum antibody or genomic DNA of N. caninum in fetal pleural fluid or sera.7

NECROPSY FINDINGS

There is no treatment that can be used to curtail an ongoing abortion epidemic. Possible drug therapies are generally not considered an option because of likely unacceptable milk and meat residues and withdrawal problems. DIFFERENTIAL DIAGNOSIS Serology and/or polymerase chain reaction can confirm infection in individual cows. Because of the high prevalence of infection, and the occurrence of congenital infection, care must be taken in extrapolating the results of a single positive diagnosis to problems of abortion. The high rate of natural congenital infection means that evidence of infection in an aborted fetus is not proof of causation of abortion, and fetal examination should be coupled to serologic examination of aborting and nonaborting animals in the herd to assess statistical differences. • Other causes of abortion in cattle • Weak calf syndrome

Goodswen SJ, Kennedy PJ, Ellis JT. A review of the infection, genetics, and evolution of Neospora caninum: from the past to the present. Infect Genet Evol. 2003;13:133-150. Gondim LF. Neospora caninum in wildlife. Trends Parasitol. 2006;22:247-252. Hemphill A, Vonlaufen N, Naguleswaran A. Cellular and immunological basis of the host-parasite relationship during infection with Neospora caninum. Parasitology. 2006;133:261-278. Innes EA. The host-parasite relationship in pregnant cattle infected with Neospora caninum. Parasitology. 2007;134:1903-1910. Innes EA, Bartley PM, Maley SW, Wright SE, Buxton D. Comparative host-parasite relationships in ovine toxoplasmosis and bovine neosporosis and strategies for vaccination. Vaccine. 2007;25:5495-5503. Williams DJ, Hartley CS, Björkman C, Trees AJ. Endogenous and exogenous transplacental transmission of Neospora caninum—how the route of transmission impacts on epidemiology and control of disease. Parasitology. 2009;136:1895-1900.

REFERENCES 1. Radostits O, et al. Diseases associated with protozoa. In: Veterinary Medicine: A Textbook of the

1819

Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1509. 2. Dubey JP, Lindsay DS. Vet Clin North Am Food Anim Pract. 2006;22:645. 3. Dubey JP, Schares G. Vet Parasitol. 2011;180:90. 4. Dubey JP, et al. Clin Microbiol Rev. 2007;20:323. 5. Reichel MP, et al. Int J Parasitol. 2013;43:133. 6. Dubey JP, et al. J Comp Pathol. 2006;134:267. 7. Dubey JP, Schares G. Vet Parasitol. 2006;140:1. 8. Innes EA, Vermeulen AN. Parasitology. 2006;133(suppl):S145. 9. Reichel MP, Ellis JY. Int J Parasitol. 2009;39:1173. 10. Schlafer DH, Miller RB, Maxie MG, eds. Female genital system. In: Jubb, Kennedy and Palmer’s Pathology of Domestic Animals. Vol. 3. 5th ed. Edinburgh, UK: Saunders; 2007:429.

DOURINE (MALADIE DU COIT) SYNOPSIS Etiology Trypanosoma equiperdum. Epidemiology Venereal disease of horses, mules, and donkeys, endemic in southern and northern Africa, Asia, and possibly South and Central America. Clinical signs Primary genital signs, secondary cutaneous signs, and tertiary nervous signs and emaciation. Lesions Edematous swelling and later, depigmentation of external genitalia, emaciation, anemia, and subcutaneous edema. Differential diagnosis list • Nagana • Surra • Coital exanthema • Equine infectious anemia. • Purulent endometritis Treatment Chronic cases unresponsive to trypanocides and may become carriers. Treatment is thus not recommended. Control Elimination of reactors, control of breeding and movement of animals in affected regions or countries.

ETIOLOGY

Trypanosoma equiperdum belongs to the brucei group, subgenus Trypanozoon, but occurs only as long, slender, and monomorphic form. It may be more appropriately referred to as T. brucei equiperdum. Unlike T. brucei. brucei, it has lost part of its kinetoplast DNA (hence dyskinetoplastic). The parasite is morphologically indistinguishable from T. evansi in blood smears. T. equiperdum is the only pathogenic trypanosome that does not require an arthropod vector for its transmission. It resides more in extra vascular tissue fluid than in blood.

EPIDEMIOLOGY Occurrence Dourine is endemic in Asia, Africa, southeastern Europe, and Central America. It has been eradicated from North America, and strict control measures have reduced the

1820

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

incidence to a low level in most parts of Europe. It occurred in Italy in 2011.1 The disease is endemic in parts of Ethiopia and Namibia and is rarely reported in other parts of sub-Saharan Africa. It has not been reported in Latin America for over 20 years. It is possible that lack of reporting in some countries may be caused by very strict international regulations that tend to discourage official notification of the disease. All Equidae are susceptible, and natural infection is known to occur only in horses, mules, and donkeys. In Ethiopia, the disease is more prevalent during the breeding season from June to September.2 Measures of Disease Occurrence In most countries, dourine now occurs only sporadically; its prevalence has declined generally because the horse is no longer that important militarily, economically, and agriculturally, and because of strict control measures in many countries. A recent survey of 237 horses from an endemic area of Ethiopia showed that infection rates varied with the method of examination.3 The rates were 4.6% based on standard parasitologic methods, 27.6% on serology, and up to 47.6% on DNA detection by PCR. This was the first time in more than 30 years that a fresh strain of T. equiperdum was isolated from clinical cases of dourine. Case mortality varies; in Europe, it may be as high as 50% to 70%, but it is much lower elsewhere, although many animals may have to be destroyed as a means of control. Methods of Transmission Natural transmission occurs only by coitus, but infection can also be acquired through intact oral, nasal, and conjunctival mucosae in foals at birth. The source of infection may be an infected stallion or mare actively discharging trypanosomes from the urethra or vagina, or an uninfected male acting as a physical carrier after serving an infected mare. The trypanosomes inhabit the urethra and vagina but disappear periodically so that only a proportion of potentially infective matings result in infection. Invasion occurs through intact mucosa, and no abrasion is necessary. Risk Factors T. equiperdum is incapable of surviving outside the host. Like other trypanosomes, it also dies quickly in cadavers. Some animals, especially donkeys and mules, may be clinically normal but act as carriers of the infection for many years. Because the disease does not require an arthropod vector for its transmission, and in view of the extensive movement of horses across continents that now takes place, the risk of infection, though small, is present in every country, as with any other venereal disease. Thoroughbred horses are more susceptible than indigenous horses, and donkeys tend to show more chronic signs.

Immune Mechanisms Infected animals produce antibodies to successive antigenic variants, as in T. brucei. Recovered animals often become carriers. Blood from infected horses is rarely infective to other horses, and the disease is not easily transmitted to ruminants under experimental conditions. Humans are not affected. Biosecurity Concerns There are none except when animals have to be moved internationally.

PATHOGENESIS

T. equiperdum shows a remarkable tropism for the mucosa of genital organs, the subcutaneous tissues, and the peripheral and CNSs. Trypanosomes deposited during coitus penetrate the intact genital mucosa, multiply locally in the extracellular tissue space, and produce an edematous swelling that may later undergo fibrosis. Subsequent systemic invasion occurs, and localization in other tissues causes vascular injury and edema, manifested clinically by subcutaneous edema. Invasion of the peripheral nervous system and the spinal cord leads to incoordination and paralysis.

CLINICAL FINDINGS

The severity of the clinical syndrome varies depending on the strain of the trypanosome and the general health of the horse population. The disease in Africa and Asia is much more chronic than in South America or Europe and may persist for many years, often without clinical signs, although these may develop when the animals’ resistance is lowered by other disease or malnutrition. The incubation period varies between 1 and 4 weeks, but could extend to more than 3 months in some animals. Initial signs may not be recognized until the breeding season. The ensuing disease will manifest genital signs in the primary stage, cutaneous signs in the secondary stage, and nervous signs in the tertiary stage. In stallions, the initial signs are swelling and edema of the penis, scrotum, prepuce, and surrounding skin, extending as far forward as the chest. Paraphimosis may occur, and inguinal lymph nodes are swollen. There is a moderate mucopurulent urethral discharge. In mares, the edema commences in the vulva and is accompanied by a profuse fluid discharge, hyperemia, and sometimes ulceration of the vaginal mucosa. The edema spreads to the perineum, udder, and abdominal floor. In Europe, the disease is more severe; genital tract involvement is often accompanied by sexual excitement and more severe swelling. In the secondary stage, cutaneous urticaria-like plaques, 2 to 5 cm in diameter, develop on the body and neck and disappear within a few hours up to a few days. These so-called silver dollar spots are pathognomonic for dourine but are not always present

and are uncommon in endemic areas. Succeeding crops of plaques may result in persistence of the cutaneous involvement for several weeks. Progressive anemia, emaciation, weakness, and nervous signs that appear at a variable time after genital involvement characterize the tertiary stage. Stiffness and weakness of the limbs are evident and incoordination develops, progressing terminally to ataxia and paralysis. Marked atrophy of the hindquarters is common, and in all animals there is loss of condition, in some to the point where extreme emaciation necessitates destruction. Lack of coordination of the hind legs, swelling of the external genitalia, and emaciation were the most common clinical signs in horses suspected to have dourine in Ethiopia.

CLINICAL PATHOLOGY

Trypanosome detection is difficult, but should be attempted in edema fluid, subcutaneous plaques, and vaginal or urethral washings or blood in early stages. Inoculation of blood into laboratory rodents is not as helpful as with other members of the brucei group. An efficient CFT is available and was the basis for a successful eradication program in Canada. However, the test does not distinguish between members of the brucei group. Other serologic tests that can be used include the IFAT, the capillary agglutination test for trypanosomes, and the ELISA, but the CFT remains the most reliable. Serologic tests do not distinguish between members of the brucei group; hence they are of limited value in areas where T. brucei or T. evansi is endemic, even when monoclonal antibodies are used. In recent interlaboratory ring trials to evaluate serologic methods for dourine diagnosis, 9 out of 22 laboratories observed a false-positive result with a known T. evansipositive serum, whether by CFT or IFAT.4 However, diagnosis can be made based on serologic tests and characteristic clinical signs under the right epidemiologic setting.2 PCR has been used to detect trypanosome DNA and is an indication of an active infection, unlike serologic tests that detect past and current infections. Still, the PCR test cannot yet distinguish T. equiperdum from T. evansi or T. brucei.5,6 With the recent isolation of new strains of T. equiperdum from clinical cases in Ethiopia,3 the first in 4 decades worldwide, there is hope that new internationally recognized tests for the diagnosis of dourine will be developed soon.

NECROPSY FINDINGS

Emaciation, anemia, and subcutaneous edema are always present, and edema of the external genitalia may be evident or the external genitalia may have healed, leaving the characteristic depigmented scars of permanent leukodermic patches. Lymph nodes

Toxic Agents Primarily Affecting the Reproductive System

VetBooks.ir

are enlarged, and there is softening of the spinal cord in the lumbosacral region. Histologic lesions consist of lymphoplasmacytic infiltration in the spinal nerves, ganglia, and meninges of the lumbar and sacral regions and in affected skin and mucosa. Trypanosomes can be found in sections of the skin and genital mucosa during the primary and secondary phases of the infection. Affected lymph nodes show nonspecific lymphoid hyperplasia. DIFFERENTIAL DIAGNOSIS The full clinical syndrome is diagnostic, when present, because no other disease has the clinical and epizootiologic characteristics of dourine. However, when the full clinical picture is not developed, other diseases like nagana, surra, coital exanthema, equine infectious anemia, and purulent endometritis should be considered. With one exception, all recent reports of the disease have been based on clinical signs, serology, and detection of trypanosome DNA, but not on parasitologic detection.

TREATMENT TREATMENT AND CONTROL None is recommended.

Many trypanocidal drugs have been used in the treatment of dourine, but results are variable, chronic cases in particular are unresponsive to treatment. The main drawback is that treated animals may remain inapparent carriers and could continue to spread the disease or complicate serologic tests. Nevertheless, in Ethiopia, treatment of experimentally infected horses with Cymelarsan at 0.25 mg/kg BW was found to be effective for both acute and chronic cases.7 Berenil (diminazene) at 7 mg/kg BW as a 5% solution injected IM, with a second injection of half the dose 24 hours later, or suramin (10 mg/kg IV for two to three treatments at weekly intervals), or quinapyramine sulfate (3–5 mg/kg in divided doses injected subcutaneously) have been tried in the past.

CONTROL

In dourine-free countries, an embargo should be placed on the importation of horses from countries in which the disease is endemic, unless the animals have been properly tested and found negative. Eradication on an area or herd basis is by the application of the CFT, along with strict control of breeding and movement of horses. Positive reactors are disposed of, and two negative tests not less than a month apart can be accepted as evidence that the disease is no longer present. Castration or neutering of infected

animals is not adequate because mating can still occur. FURTHER READING

Abebe G. Trypanosomosis in Ethiopia. Ethiopia J Biol Sci. 2005;4:75-121. Barrowman P, et al. Dourine. In: Coetzer JAW, Thomson GR, Tustin RC, eds. Infectious Diseases of Livestock With Special Reference to Southern Africa. Vol. 1. Cape Town: Oxford University Press; 1994:206-212. Desquesnes M. Livestock Trypanosomoses and Their Vectors in Latin America. Paris: OIE (World Organization for Animal Health); 2004. Hunter AG, Luckins AG, Trypanosomosis. In: Sewell MMH, Brocklesby DW, eds. Handbook on Animal Diseases in the Tropics. 4th ed. London: Bailliére Tindall; 1990:204-226. Luckins AG, et al. Dourine. In: Coetzer JAW, Tustin RC, eds. Infectious Diseases of Livestock. Vol. 1. 2nd ed. Cape Town: Oxford University Press; 2004:297-304. OIE. Manual of Diagnostic Tests and Vaccines for Terrestrial Animals. Vol. 2. 6th ed. 2008:845-851. Stephen LE. Trypanosomiasis: A Veterinary Perspective. Oxford: Pergamon Press; 1986.

REFERENCES

1. Pascucci I, et al. Vet Parasitol. 2013;193:30. 2. Hagos A, et al. Proceedings of ISCTRC. Kampala, Uganda: 2009:317. 3. Gari FR, et al. Trop Anim Health Prod. 2010;42:1649. 4. Cauchard J, et al. Vet Parasitol. 2014;205:70. 5. Li FJ, et al. Mol Cell Probes. 2007;21:1. 6. Tran T, et al. Parasitology. 2006;133:613. 7. Hagos A, et al. Vet Parasitol. 2010;171:200.

Toxic Agents Primarily Affecting the Reproductive System ESTROGENIC SUBSTANCES ETIOLOGY

Poisoning occurs either accidentally or intentionally from administration of a number of different products. Supplementation may be by addition to the feed, but is usually by subcutaneous implants. Many of them are used as growth promotants to increase weight gain and feed efficiency in animals.1 Estrogen in some form can be found in the following four categories of growth promotants: • Endogenous hormones (estradiol-17-β, progesterone, testosterone)1,2 • Synthetic hormones (ethinylestradiol, others)1 • Xenobiotics (zearalenone [α-zearalanol; zeranol], trenbolone)1,3 • Miscellaneous (diethylstilbestrol and related compounds such as hexestrol and dienestrol)1

EPIDEMIOLOGY Occurrence Poisoning by estrogenic substances occurs in the following circumstances: • Natural substances such as genistein present in plants and as zearalenone in fungi1,3

1821

• Dietary supplements for fattening cattle1 • Overdosage of medications used in clinical infertility cases • Pigs fed hexestrol implants in capon necks • Cattle fed on chicken litter from farms on which estrogens are used as supplements. Risk Factors Animal Risk Factors Steers implanted with an estrogen at a standard dose rate may respond in an exaggerated manner and show signs of toxicity. Estradiol implants are reputed to be associated with more of these problems than zeranol. Environmental Risk Factors Estrogens from treated animals are found in the environment in water and animal manure and may act as endocrine disrupters. Water treatment plants are able to remove most of the estrogens, but animal manure is not regulated in the many parts of the world unless it is discharged into a water supply.4-6 Farm Risk Factors Pasture may be contaminated by manure from cattle treated orally or by subcutaneous implants with estrogenic substances that pass significant amounts in the feces.2,6 Ensilage made from the pasture may also be contaminated. Human Risk Factors Estrogenic substance administration as a management tool is regarded unfavorably in many countries because of the risk of intoxication occurring in humans eating contaminated meat. Their use is banned in some and strictly controlled in others. In one small study, a palpable mammary tumor was observed in a rat implanted with a 12-mg zeranol pellet.3 The presence of environmental zearalenone has been proposed as a link to early puberty and anabolic growth effects in young girls.7

PATHOGENESIS

Signs and lesions are the direct result of amplification of the pharmacologic effects of the substances.

CLINICAL SIGNS Idiopathic Female Estrogenism In addition to the toxic effects associated with estrogens in specific plants, increased estrogenic activity is also encountered in mixed pasture, generally only at certain times and on particular fields. Clinically the effects are those of sterility, some abortions, swelling of the udder and vulva in pregnant animals and virgin heifers, and endometritis with a slimy, purulent vaginal discharge in some animals. Estrous cycles are irregular. In milking cows, there is depression of the milk yield, reduction in appetite, and an increase in the cell count of the milk.

1822

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

Male Estrogenism Steers in feedlots may exhibit excessive mounting by other steers, sometimes to the point of causing death. Head injuries caused by head-to-head butting, frequent bawling, stampedes, and pawing the ground to the point of hole-digging are other reported signs. These problems tend to pass off after a short time. Preputial prolapse may be a problem in Bos indicus cattle. Experimental feeding of zeranol to young bulls is associated with retardation of testicular and epididymal development. Nymphomania in Cows Larger doses of stilbestrol, usually administered accidentally to cows, may be associated with prolapse of the rectum and vagina and elevation of the tail head caused by relaxation of the pelvic ligaments. Susceptibility to fracture of the pelvic bones and dislocation of the hip are common sequelae. Nymphomaniac behavior in such animals results in other skeletal injuries, especially fracture of the wing of the ilium. Swine Estrogenism Common clinical signs include weight loss, decreased feed efficiency, straining, prolapse of the rectum, incontinence of urine, anuria, and death.8 Estrogens such as zearalenone ingested by sows after day 11 to 13 of the estrous cycle can be associated with retention of corpora lutea and a syndrome of anestrus or pseudopregnancy, which typically persists for 45 to 60 days postestrus. This effect may occur at zearalenone concentrations of 3 to 10 ppm in the diet. Pregnant sows given zearalenone postbreeding may have failure of implantation and early fetal abortion. Urethral Obstruction Heavy mortalities have occurred in feeder lambs after the use of implants of estrogens as a result of prolapse of the rectum, vagina, and uterus, together with urethral obstruction by calculi. The calculi consist largely of desquamated epithelial and inflammatory cells that form a nidus for the deposition of mineral; the desquamation is probably stimulated by the estrogen. Also, urethral narrowing caused by the estrogen facilitates complete obstruction by the calculi.

CLINICAL PATHOLOGY

High blood levels of estrogens are characteristic. In swine, the syndrome of anestrus associated with zearalenone will be accompanied by elevated progesterone concentrations caused by the retention of corpora lutea.

NECROPSY FINDINGS

Enlargement and vascular engorgement of accessory sex organs, especially in neutered animals, are characteristic. Uterine enlargement and keratinization of vaginal

epithelium may be detected, and in mature female swine there may be persistent multiple retained corpora lutea. Swine also show inflammation and necrosis of the rectal wall, enlargement of the kidneys, thickening of the ureters and distension of the bladder, and gross enlargement of the prostate and seminal vesicles. Histopathology on jejunum obtained from pigs treated with low doses of zearalenone and T-2 toxin showed normal crypts and villi but decreased numbers of goblet cells and acidophilic granulocytes in the mucous membrane and numerous plasma cells in the intestinal epithelium.8 FURTHER READING Adams NR. Detection of the effects of phytoestrogens on sheep and cattle. J Anim Sci. 1995;73:1509-1515. Burnison BK, Hartman A, Lister A, et al. A toxicity identification evaluation approach to studying estrogenic substances in hog manure and agricultural runoff. Environ Toxicol Chem. 2003;22:2243-2250. Leffers H, Naesby M, Vendelbo B, et al. OEstrogenic potencies of zeranol, oestradiol, diethylstiboestrol, bisphenol-A and genistein; implications for exposure assessment of potential endocrine disruptors. Hum Reprod. 2001;16:1037-1045. Soto AN, Calabro JM, Prechtl NV, et al. Androgenic and estrogenic activity in water bodies receiving cattle feedlot effluent in Eastern Nebraska, USA. Environ Health Persp. 2004;112:346-352.

REFERENCES

1. Biswas S, et al. J Soil Water Con. 2013;66:325. 2. Khanal SK, et al. Environ Sci Technol. 2006;40: 6537. 3. Zhong S, et al. Anticancer Res. 2011;31:1659. 4. Chen TS, et al. Sci Total Environ. 2010;408:3223. 5. Alvarez DA, et al. Water Res. 2013;47:3347. 6. Gadd JB, et al. Environ Pollut. 2010;158:730. 7. Massart F, et al. J Pediatr. 2008;152:690. 8. Andretta I, et al. Arch Zootech. 2010;59:123.

PHYTOESTROGEN TOXICOSIS SYNOPSIS Etiology Ingestion of plants that produce estrogen (phytoestrogens) resulting in a number of reproductive problems. Epidemiology Pastures dominated by specific strains of legumes, in lush growth mode, or hay or silage made from such pasture, are associated with problems if exposure is prolonged. Sheep are much more susceptible than cattle. Clinical pathology Positive estrogen assay in blood. Lesions Live animals: Severe flock infertility in sheep; prolongation of estrus periods, interestrus periods shortened. Postmortem: Ewes show cystic endometrial degeneration. Diagnosis confirmation Laboratory assay of feed, blood, and tissue; the appearance of genital pathology at necropsy, or with a uterine biopsy or laparoscopy.

Treatment None. Control Grazing management, use of low-phytoestrogen cultivars.

ETIOLOGY Important estrogenic substances found in plants and fungi include the following: • Plants • Coumestans (coumestrol, 4-methoxycoumestrol, repensol, trifoliol)1 • Isoflavones (daidzein, formononetin, genistein, biochanin A, glycitein)2,3 • Isoflavan (equol, a metabolite of daidzein)3 • Fungi (resorcylic acid lactones [zearalenone])4 Compared with pharmaceutical agents, these substances have low estrogenic activity, but they are associated with serious clinical effects because of the high concentrations they reach in some plants and daily intake over long periods. The coumestans are most common in plants of the Medicago genus; isoflavones are most common in the Trifolium, Baptisia, and Cytisus genera. Only Medicago and Trifolium spp. are of any importance to animals. Those likely to contain sufficient amounts to be associated with disease are Fusarium (variety of species); contains zearalenone4 Glycine max (soybean; contains coumestans and isoflavones; affects pigs) Medicago sativa (alfalfa, lucerne; contains coumestans; affects cattle, sheep) Trifolium alexandrinum (isoflavones) T. alpestre (alpestrine clover; contains isoflavones) T. pratense (red clover; contains isoflavones; affects sheep)1 T. repens (white clover, Ladino clover; contains coumestans)1 T. subterraneum (subterranean clover; contains isoflavones; affects sheep).

EPIDEMIOLOGY Occurrence Animals on pasture are at the greatest risk, but poisoning can also occur on diets containing prepared feeds such as soybean (Glycine max) meal, or moldy feed containing Fusarium fungi. Risk Factors Animal Factors Phytoestrogen toxicosis is clinically important only in sheep. Cattle are generally considered to be less sensitive than sheep.1,5,6 For example, cows can ingest large amounts of estrogens (over 40 g per day per cow) in red clover without showing any reduction in reproductive efficiency. Horses usually graze the toxic pasture without ill effects.

Toxic Agents Primarily Affecting the Reproductive System

VetBooks.ir

Massive reproductive wastage has been experienced in sheep on pastures dominated by such plants as Trifolium subterraneum, and the death rate from dystocia and prolapse of the uterus can also be high. The most common abnormality is a failure to conceive, even with multiple matings, and the flock breeding status worsens progressively, with the lambing percentage falling from a normal 80% down to 30%. Sheep eating a lot of estrogenic clover in the spring can become temporarily infertile, but are normally fertile again by the usual breeding season in the autumn. However, ingestion of the plant in several successive years is associated with “permanent clover disease”—infertility from which ewes do not recover. Under these conditions sheep farming becomes unprofitable, and large areas of country have been made unsuitable for sheep raising because of this disease. Human Factors Various phytoestrogens have been found in foods of animal origin (eggs, milk, meat, fish, and seafood). Equol was found in several foods, including eggs, milk, and meat.7 Not all phytoestrogens are harmful and many of them are have known human health benefits.8 Many, however, are endocrine disruptors, which means that they can produce adverse health effects as well. Plant Factors The estrogenic activity of pastures depends on the degree of domination of the pasture by the toxic plants, the variety of the plant species, and the duration of the animal’s exposure to them. Newly sown pastures are usually most toxic because of domination by the sown legume. Pastures deficient in phosphorus are also likely to be clover dominant. High nitrogen fertilizer applications reduce phytoestrogen content. Varieties of Trifolium subterraneum, e.g., Yarloop, Dwalganup, Dinninup, and Geraldton, are much more toxic than Bacchus Marsh and Daliak. Pastures containing more than 30% of the first four varieties are likely to be unsafe. In some clovers, e.g., red clover, the estrogen content varies with the season, and is high in early spring, low in midsummer, and high again in the autumn after the hay has been taken off. Insect damage to pasture can increase the estrogen content 10-fold, and bacterial infection (e.g., by Pseudopezzia medicaginnis, a leaf-spotting organism on alfalfa) and fungal infection by 100-fold. Plants that have matured in the field and set seed have no estrogenic potency, but the making of potent fodder into hay causes little depression of estrogen content. Clover ensilage can contain high levels of estrogens, and the ensiling process is considered to increase the estrogenic effect of clover 3- to 5-fold. Trifolium repens (white clover, in contradistinction to Ladino clover), does not have

a high content of estrogens.1 However, when heavily infested with fungi it can contain significant amounts. It is thought that the production of estrogens is a byproduct of the plant’s mechanism of resistance to the fungal infection. Ladino clover, a largegrowing variety of white clover, may contain large quantities of a highly active estrogen (coumestrol), and when it dominates a pasture and is grazed when the pasture is lush, it may be associated with the cornification of vaginal epithelium and functional infertility in ewes. Three estrogenic compounds have been isolated from T. pratense (red clover), and where this plant dominates the pasture a clinical syndrome similar to that associated with subterranean clover may be observed. Ewes grazing on red clover pasture, especially a toxic cultivar of the plant, may have their conception rate at the first mating cycle reduced from 75% to as low as 25%.

PATHOGENESIS

Much of the metabolism of phytoestrogens in ruminants occurs in the rumen as well as in the liver.1 The differences between sheep and cattle in the ruminal metabolism of these compounds are thought to be the reason for the comparative freedom of cattle from the clinical disease. The amount of phytoestrogen ingested by a ewe on a highly poisonous pasture may equal her daily estrogen secretion at the peak of her estrous cycle. The effect of the phytoestrogens is exerted mainly on the uterus and ovaries. Structurally, there is hyperplasia and hypertrophy of the epithelium of the uterus, vagina, and cervix, and dysplasia of the granulosa cells of the ovary, with a consequent reduction in secretion of estradiol. Increases in teat size and milk secretion are additional, secondary effects. The functional abnormality is not one of estrus; in sheep the demonstration and duration of estrus may be normal or depressed, and the defect is one of sperm transport because of changes in the composition of cervical mucus and the structure of cervical glands. The change is to more watery mucus, and this is the basis of a test in affected sheep in which the watery mucus is more readily absorbed by a cottonwood plug inserted in the vagina. The increased weight of the plug is a positive test. It is possible that a good deal of the infertility seen in ewes on improved clover pasture may be associated with its high estrogen content, in spite of the absence of the more dramatic evidence of hyperestrogenism described earlier. Because it is necessity to use this pasture, a great deal more needs to be known about the seasonal occurrence of the estrogenic substances and the management of sheep grazing the pasture so that the effects of the disease can be minimized.

1823

CLINICAL FINDINGS Ewes Clover disease, the severe clinical manifestation of phytoestrogen poisoning, and rarely seen today, includes dystocia, prolapse of the uterus or vagina, severe infertility, and death. The more common and less severe field expression of phytoestrogen poisoning is a significant decrease in fertility rate. It may be temporary with normal reproductive efficiency returning soon after the ewes are moved to clover-free pasture. In ewes exposed to a low level intake of estrogens over a long period, e.g., in excess of two grazing seasons, a process of irreversible “defeminization” may occur. This is a state of permanent subfertility. The estrous cycle is normal, but an abnormally large number of ewes fail to conceive. In affected flocks, there may also be a high incidence of maternal dystocia caused by uterine inertia, or failure of the cervix or vagina to dilate. Affected ewes show little evidence of impending parturition and many full-term fetuses are born dead. Male Castrates Wethers may secrete milk, and metaplasia of the prostate and bulbourethral glands is evident. These can be detected at an early stage of development by digital rectal palpation. Continuing hyperplasia and cystic dilatation of these glands is associated with their prolapse in a subanal position, followed by rapid weight loss and fatal rupture of the bladder. Rams usually show no clinical abnormality, and their fertility is not impaired. Cattle exhibit clinical signs less often than sheep, with experimental reports of decreases in conception and fertilization caused by prolongation of oocyte maturation and decreased sensitivity of the corpus luteum to luteolytic agents.5,6 Temporary infertility; discharge of cervical mucus; and swelling of the mammary gland, vulva, and uterus have all been recorded in cattle. Gilts exposed to genistein may develop structural changes and abnormalities in the cervix and uterus.9

CLINICAL PATHOLOGY

Laboratory assays are available and essential to diagnosis and monitoring of feed contents of phytoestrogens.7 Chemical assays are not as sensitive as biologic assessments based on increased size of genitalia in subject animals.

NECROPSY FINDINGS

Severe cystic degeneration of the endometrium is present in the most severe cases. Similar clinical and histopathologic changes have been produced by the daily injection of 0.03 mg of diethylstilbestrol per ewe for a period of 6 months. There is also a long-term change in the cervix with an increased incidence of cervicitis and a histologically observable transformation to a uterine-like

1824

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

appearance. In ewes on a long-term intake of toxic pasture, the lesions include elevation of the tail head, partial fusion of the vulvar labia, and clitoral hypertrophy. Diagnostic confirmation of phytoestrogen poisoning requires laboratory assay of feed, blood, and tissue, and the appearance of genital pathology at necropsy, or with a uterine biopsy, or laparoscopy. DIFFERENTIAL DIAGNOSIS Differential diagnosis list • Overdose of pharmaceutical preparation as part of a program to improve fertility in a herd. • Overdose of an implant or feed additive with a growth stimulant that has estrogenic capability.

TREATMENT Administration of testosterone is a logical response to poisoning but appears to be an unlikely commercial proposition.

CONTROL

Avoidance of high estrogenic activity strains of the respective plants, grazing management to avoid dangerous pasture at the most toxic part of the season, and dilution of the estrogen intake by providing additional and alternative feeds, are all used to control the disease. Prevention of clover disease can only be achieved by proper management of sheep and pasture to avoid ingestion of excessive amounts of estrogens. Vaccination with a phytoestrogen-immunogenic protein conjugate has produced good levels of antibodies, but has not been successful in preventing the problem. Careful management of flocks on estrogenic pasture can significantly improve reproductive output. FURTHER READING Adams NR. Detection of the effects of phytoestrogens on sheep and cattle. J Anim Sci. 1995;73:1509-1515. Hughes CL. Phytochemical mimicry of reproductive hormones and modulation of herbivore fertility by phytoestrogens. Environ Health Persp. 1988;78:171. Kuiper G, et al. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor β. Endocrinology. 1998;139:4252-4263. Radostits O, et al. Phytoestrogen poisoning. In: Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1873.

REFERENCES

1. Steinshamn H, et al. J Dairy Sci. 2008;91:2715. 2. Hoikkala A, et al. Mol Nutr Food Res. 2007;51:782. 3. Jackman KA, et al. Curr Med Chem. 2007;14:2824. 4. Zinedine A, et al. Food Chem Toxicol. 2007;45:1. 5. Borzym E, et al. Med Weter. 2008;64:1107. 6. Piotrowska KK, et al. J Reprod Dev. 2006;52:33. 7. Kuhnle GGC, et al. J Agric Food Chem. 2008;56:10099. 8. Patisaul HB, et al. Front Neuroendocrinol. 2010;31:400. 9. Ford JA, et al. J Anim Sci. 2006;84:834.

ZEARALENONE TOXICOSIS SYNOPSIS Etiology Zearalenone is an estrogenic mycotoxin produced primarily by fungus in the genus Fusarium, which is the causative agent. F. graminearum is the species most responsible for animal reproductive problems, but F. cerealis, F. culmorum, F. cookwellense, F. equiseti, and F. semitectum are contaminants of moldy maize, wheat, oats, and barley grain and cause issues as well. Epidemiology Global issue with zearalenone found in a variety of cereals and foodstuffs in many countries. Clinical pathology None in particular; progesterone levels may be decreased. Lesions Associated with hyperestrogenism and include abortions, stillbirths, mammary gland enlargement and secretions, vulvar edema, and vaginitis in females as well as testicular atrophy and mammary gland enlargement in males. Diagnostic confirmation Presence of zearalenone and/or metabolites in feces, urine, and serum; presence in feedstuffs. Treatment Remove animals from contaminated feed and correct prolapses. Control Keep moisture content of stored grain below 15%–16%; feed contaminated grains to less susceptible animals.

ETIOLOGY Zearalenone is a nonsteroidal estrogenic mycotoxin produced primarily by fungi in the genus Fusarium. F. graminearum is the species most responsible for animals’ reproductive problems, but F. cerealis, F. culmorum, F. cookwellense, F. equiseti, and F. semitectum are contaminants of moldy maize, wheat, oats, and barley grain and are associated with toxicosis.1,2 Swine are most commonly affected, but cases have occurred in sheep and cattle3,4 and more rarely in horses.5

EPIDEMIOLOGY Occurrence The fungi that produce zearalenone primarily colonize corn, but they also infect other cereal grains such as barley, wheat, and oats.1,2 Zearalenone has also been detected in a number of other plants including rice, sorghum, millet, and soybeans. Most typically, contamination occurs from high moisture during storage; field contamination has been reported but occurs less often. Zearalenone has been detected in pastures in New Zealand, which has been associated with infertility in ewes.6 Contamination of food and animals is considered a global problem because zearalenone has been found in Africa, Asia, Australia, Europe, North America, and South America.2

Risk Factors Animal Risk Factors Swine of all ages, but especially prepubertal gilt, are the most sensitive to the effects of zearalenone. The primary effects are reproductive and depend on the dose and time of administration in relationship to the animal’s estrous cycle.5,6 Farm Risk Factors Elevated levels of zearalenone in the feed are primarily associated with improper storage and not contamination in the field.2 Human Risk Factors There is considerable concern that humans, especially young girls, will be adversely affected by zearalenone in cereal products, milk and milk-based products, and meats. In Europe, 32% of mixed cereal samples from nine countries were found to be contaminated with zearalenone. Zearalenone is excreted in milk and present in some concentration in meats in animals with high intake, but currently the risk to humans is thought to be low.2

PATHOGENESIS

Zearalenone is rapidly absorbed following an oral exposure, with an estimated uptake of 80% to 85%.1,2 In swine, it can be detected in the serum within about 30 minutes after ingestion.2 Distribution is primarily to the adipose tissue and the ovary and uterus. The liver is the main site of metabolism, but other tissues such as the intestine, kidney, ovary, and testis are metabolic sites.1 Two different biotransformation pathways have been proposed and likely play a role in the susceptibility `of different species.1,5 Zearalenone is either conjugated with glucuronic acid or hydroxylated to α- and β-zearalenol.1,5 In swine, the preferred route is conjugation with conversion to primarily α-zearalenol.1,4,5 Sheep are similar to swine but cattle convert to β-zearalenol, a less estrogenic metabolite.4 Excretion is biliary in most species with significant enterohepatic recirculation occurring.1 Zearalenone crosses cell membranes and binds to cytosolic 17β-estradiol receptors. Once this occurs, it is translocated into the nucleus where it binds to estrogenresponsive elements and stimulates mRNA synthesis resulting in estrogen-like effects.1,3

CLINICAL FINDINGS Swine Pigs of all ages are affected, including piglets nursing on sows, which themselves show no signs of estrogenism. The most significantly affected are the 6 to 7-month-old gilts. Vulvovaginitis, including swelling of the vulva to three to four times normal size, enlargement of mammary glands, a thin catarrhal exudate from the vulva, and increased size and weight of the ovaries and uterus, is the severest form

Toxic Agents Primarily Affecting the Reproductive System

VetBooks.ir

of the poisoning.3,6 Prolapse of the vagina is common (up to 30% of affected pigs) and there is prolapse of the rectum in some pigs (5%–10%). The toxin reduces serum progesterone levels in sows, but the administration of progesterone to affected gilts does not counteract the estrogenic effects. The syndrome is indistinguishable from that produced by long-term overdosing with diethylstilbestrol. Signs appear 3 to 6 days after feeding of moldy grain commences and disappear soon after the feeding stops. The mortality rate is high because of the secondary development of cystitis, uremia, and septicemia. The more important manifestation of the poisoning may be infertility, including absence of estrus, high levels of stillbirth, neonatal mortality, and reduced litter size. Small fetal size, fetal malformations, splayleg and hindlimb paresis, pseudopregnancy, and constant estrus are also recorded.3 Zearalenone in male pigs can induce feminizing characteristics; suppress libido; and decrease spermatogenesis, testicular weights, and serum testosterone concentrations.2 Ruminants In cattle, the effect of zearalenone is largely on conception rate, and the rate of services per conception may rise, but the overall effect is less than in sows. Milk production may be decreased.2 Behavioral estrus occurs at times unrelated to ovarian cycles and in late pregnant cows. There is idiopathic vaginitis. Symmetric enlargement of the mammary glands is recorded in prepubertal dairy heifers feeding on fungus-infected corn. Estrogenic disturbances are also suspected in sheep. Abortion is suspected to occur, and mild vulvovaginitis and hypertrophy of the uterus are recorded. Experimental feeding of zearalenone to lactating cows and ewes does result in minor contamination of their milk sufficient to produce hyperestrogenism in a lamb sucking a poisoned ewe. Horses Zearalenone toxicosis is rarely reported in horses.1 A recent study using equine ovarian cultured granulosa cells demonstrated that zearalenone may play a role in some equine reproductive disorders.5

CLINICAL PATHOLOGY

Zearalenone and its metabolites can be identified in urine, plasma, and feces by highperformance liquid chromatography7 and in feedstuffs by liquid chromatography mass spectrometry and a rapid immunoassay.8,9 In 2003, 16 countries limited the amount of allowable zearalenone in maize and cereals; the allowable concentration varies from 50 to 1000 µg/kg depending on the country.5

NECROPSY FINDINGS

On necropsy, there are nonspecific findings other than expected changes associated

with estrogen-related reproductive tract abnormalities. These include changes in ovarian weight with decreased numbers of corpora lutea, increased dead piglets, vaginal and rectal prolapses, vulvar edema and vaginitis in females, and testicular atrophy and mammary gland enlargement in males.10

DIFFERENTIAL DIAGNOSIS Differential diagnosis list • Accidental overdose of synthetic estrogen substances • Estrogenic substances • Phytoestrogens

TREATMENT Complete recovery follows when the feeding of the affected grain is stopped and no treatment other than surgical repair of the prolapsed organs is attempted.

CONTROL

The moisture content of grains should be kept below 15% to 16% during storage. If contaminated feeds must be used, they should be fed to animals less susceptible to toxicosis. The 2006 EU guidelines for zearalenone in feeds recommend that piglets and gilts do not receive more than 0.1 mg zearalenone/kg BW; sows and fattening pigs no more than 0.25 mg zearalenone/kg BW; and sheep, goats, calves, and dairy cows no more than 0.5 mg zearalenone/kg BW.10 FURTHER READING Etienne M, Jemmali M. Effects of zearalenone (F2) on estrous activity and reproduction in gilts. J Anim Sci. 1982;55:1-10. Tanaka T, Hasegawa A, Yamamoto S, et al. Worldwide contamination of cereals by the Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone. Survey of 19 countries. J Agric Food Chem. 1988;36:979-983. Radostits O, et al. Zearalenone. In: Veterinary Medicine: A Textbook of the Disease of Cattle, Horses, Sheep, Goats and Pigs. 10th ed. London: W.B. Saunders; 2007:1911.

REFERENCES

1. Minervini F, et al. Int J Mol Sci. 2008;9:2570. 2. Zinedine A, et al. Food Chem Toxicol. 2007;45:1. 3. Kanora A, et al. Vet Med-Czech. 2009;12:565. 4. Malekineja HR, et al. Vet J. 2006;172:96. 5. Minervini F, et al. Reprod Biol Endocrinol. 2006;4: 62. 6. Upadhaya SD, et al. Asian-Aus J Anim Sci. 2010;23:1250. 7. Songsermsakul P, et al. J Chromatography B. 2006;843:252. 8. Tanaka H, et al. Rapid Commun Mass Spectrom. 2006;20:1422. 9. Kolosova AY, et al. Anal Bioanal Chem. 2007;389:2103. 10. Tiemann U, et al. Food Addit Contam. 2007;24: 306.

1825

MARE REPRODUCTIVE LOSS SYNDROME (EARLY FETAL LOSS, LATE FETAL LOSS, FIBRINOUS PERICARDITIS, AND UNILATERAL UVEITIS) SYNOPSIS Etiology Exposure to Eastern tent caterpillars (ETCs; Malacosoma americanum), in particular during the spring when the caterpillars are most active. Epidemiology Occurs primarily in the Ohio River valley, but reported in other states. Risk factors are the presence of black cherry trees on pasture, ETC, and feeding hay on the ground. Clinical pathology Culture of fetal and placental tissue most commonly results in growth of non–β-hemolytic streptococci and/or Actinobacillus. Lesions Inflammation of the intraamniotic umbilical cord (funisitis), premature placental separation, placental edema, placentitis, diffuse alveolitis, and hemorrhage in a variety of organs. Diagnostic confirmation Based on the presence of appropriate clinical signs with a history of exposure of affected horses to ETCs. Treatment Supportive care only. Control Removal of cherry trees from pasture, spraying ETC nests and pastures with pyrethrin pesticides, keeping horses off pasture or muzzling mares on pasture during active ETC months.

ETIOLOGY

In 2001 an epidemic of early fetal loss (40–80 days; range 40–140 days) and late fetal loss (about 340 days) was recognized in north central Kentucky, southern Ohio, and Tennessee affecting over 3500 mares.1,2 It occurred again in 2002 but far fewer horses were affected. The epidemic was termed mare reproductive loss syndrome (MRLS). At the same time there was also a marked increase in incidence of birth of weak foals and fibrinous pericarditis and unilateral uveitis in adult horses in the same region.1-3 Research in horses and pigs confirmed the causative agent as Malacosoma americanum, the Eastern tent caterpillar (ETC). Similar episodes of equine abortions, now referred to as equine amnionitis and fetal loss (EAFL), occurred in Australia and have been associated with the Ochrogaster lunifer, the processionary caterpillar.4,5

EPIDEMIOLOGY

Historically, many epidemiologic studies were performed to determine the source of the epidemic. Several toxins such as fescue, nitrate/nitrite, phytoestrogens, and mycotoxins were examined and ruled out leaving a

1826

Chapter 18 

■ 

Diseases Primarily Affecting the Reproductive System

VetBooks.ir

strong association between the presence of ETCs (M. americanum, black cherry trees (Prunus serotina), and feeding horses hay off the ground. Black cherry trees were involved because they are the preferred host tree for ETC and may be a source of cyanide. Black cherry trees (i.e., cyanide) were ruled out as a cause of MRLS, and an association with ETC was examined experimentally. In several different experiments, pregnant horses (50 to 200 days’ gestation) were exposed to various forms of ETC and only those mares exposed to live ETC larvae aborted. These were the first studies to reproduce MRLS and demonstrate that ETC could cause pregnancy loss in mares. Further studies demonstrated that the cuticle (setae; hairs) is the structure responsible for the abortigenic activity.1,2 Culture of the placental fluid or fetal tissues in both early and late losses showed non–β-hemolytic streptococci and Actinobacillus, which are bacteria routinely found in the oral cavity of horses.2,6 Finally, the syndrome was reproduced in pigs with abortions occurring 13 to 16 days after first ingestion.1 More important, histopathologic examination showed ETC setae imbedded in the gastrointestinal mucosa that were surrounded by microgranulomatous lesions.1,2,6 A similar pattern was subsequently confirmed in pregnant and nonpregnant mares.

obtained from pasture, and being fed exclusively in pasture during the final 4 weeks of gestation. All of these factors favor exposure to ETC. Risk factors for pericarditis include presence of mares or foals with MRLS on the farm, grazing, and exposure to ETC. Risk factors for uveitis have not been defined.

Occurrence The first well-studied and documented outbreak of abortions occurred from April 26 through mid-June of 2001, with a lower incidence of disease during the same months in 2002. An abortion storm, which may have been related, occurred in Kentucky in 1991 and 1982, but no epidemiologic studies were performed.1 In 2006, a similar syndrome associated with large numbers of ETC was reported in Florida.2 The 2001 to 2002 outbreak caused early fetal loss in 25% to 63% of mares on one-third of farms, 14% to 24% on another third, and 2% to 13% on the remaining one-third. Approximately 21% of mares pregnant at 42 days’ gestation were not pregnant when examined at 70 to 90 days’ gestation. The expected pregnancy loss rate between 42 days and parturition is 12%. Over 3500 mares (3000 early fetal losses; 500 late fetal losses) aborted during the outbreak.1,3 The economic losses incurred because of MRLS during 2001 and 2002 are estimated to be $500 million.1

PATHOGENESIS

Risk Factors Animal Risk Factors Risk factors for the disease are the presence of black cherry trees, exposure to ETC (especially the presence of large numbers of caterpillars on pasture), and pasturing or feeding hay to horses at pasture. For late-term abortion the risk factors include increased amount of time at pasture, less time in stall, feeding concentrate on the ground, increased proportion of feed

Farm Risk Factors ETCs are endemic to the eastern United States including the Ohio River valley. Egg masses are laid on many trees in the Rosaceae family including black cherry trees, which are the preferred host. Eggs hatch in the early spring when the cherry trees bud. Local populations of the caterpillars fluctuate dramatically from year to year, but mares are likely exposed to small numbers of the caterpillars every spring. Climatic conditions that favor survival of ETC and synchronize their maturation result in simultaneous hatching of large numbers of eggs. The rapid emergence of large numbers of caterpillars results in abrupt and heavy exposure of horses and consequent development of MRLS. Weather conditions thought to contribute to the 2001 outbreak include a period of low temperatures in March, above normal temperatures in April, and a frost and freeze in late April immediately followed by several warm days. The pathogenesis of the diseases associated with MRLS has not been well defined. Based on experimental studies and natural cases, ETC setae are likely involved in the pathophysiology. Two different hypotheses have been proposed: • Setae lodged in the gastrointestinal submucosa causes inflammation, form microgranulomas, and disrupt the mucosal barrier. Resident bacteria such as Actinobacillus spp. penetrate the barrier, resulting in bacteremia and hematogenous spread to the placenta, fetus, pericardium, uvea, and meninges.1,6 • Setae or parts of the exoskeleton contain an as yet unidentified toxin that is toxic to the placenta and fetus.1

CLINICAL FINDINGS Early Fetal Loss This is detected by per rectum uterine examination, either manual or using ultrasonographic visualization of uterine contents, during early pregnancy. Fetal loss occurs after 35 days, conception not being affected, and affected mares do not come into estrus because of the presence of endometrial cups, which do not regress until 100 to 180 days after ovulation.3 Mares have no clinically detectable premonitory signs of fetal loss.1,2 Ultrasonographic examination of the uterus of pregnant mares reveals that the allantoic fluid of fetuses 5 s

in highly strung breeds such as the Merino, which have a higher mismothering rate than do Romney ewes.2,3 Bonding occurs rapidly after birth, although there is some minor variation between species, with bonding starting within a few minutes of birth in sheep but taking up to 2 to 3 hours in some horses, for example. The strength of bonding also appears to vary between species. The bonding of the dam to the neonate is usually quite specific, although this can be modulated by management systems, and the neonate may be less selective and will often attempt to suck other dams. With sheep lambed under intensive lambing practices, this can lead to high rates of mismothering and subsequent abandonment, when preparturient “robber” ewes adopt lambs from multiple births. A high degree of shepherding is required to minimize loss in these management systems, whereas in extensive systems a strong bonding is established if the ewe and lamb are allowed to remain relatively undisturbed on the lambing site for 6 hours. A scoring system is available to allow objective assessment of the vigor of newborn lambs (Tables 19-1 and 19-2). There is evidence of genetic and parental (sire) effects on the ability of lambs to follow

the dam and to avoid mismothering. These effects appear to be modest. 2,4,5 Vaginal cervical stimulation and the central release of oxytocin are thought to be important in initiating maternal behavior, although caudal epidural anesthesia for delivery does not effect mothering or bonding. Sucking is also a major determinant. Recognition is olfactory and auditory and mediated by the release of neurotransmitters. Bonding is often slower with primiparous dams and is also delayed where there is postpartum pain. A failure of bonding leads to rejection and abandonment of the neonate. Maternal care is also important to neonatal survival, and there is significant difference in litter mortality from crushing and injury among sows related to sow behavior and their response to piglet distress calls. A description of normal and abnormal behavioral patterns of the mare and foal is available, and techniques for fostering have been described. REFERENCES 1. Bickell SL, et al. Anim Prod Sci. 2010;50:675. 2. Hergenhan RL, et al. Anim Prod Sci. 2014;54:745. 3. Plush KJ, et al. Appl Anim Behav Sci. 2011;134:130. 4. Brien FD, et al. Anim Prod Sci. 2014;54:667. 5. Hinch GN, et al. Anim Prod Sci. 2014;54:656.

TEETH CLIPPING OF PIGLETS It is necessary to shorten the needle teeth of the upper and lower jaw of the newborn pig using a clean pair of sharp nail clippers or a grinding wheel. It is essential to practice good hygiene or infection of tooth roots can occur, leading to local inflammation and infection with the possibility of abscessation associated with Fusobacterium and Trueperella. It is not done before 6 hours of age because it will interfere with the absorption of colostrum. It is done to prevent damage to the sow’s teats or to other piglets before 7 days after birth. Damage to the sow’s teats will cause pain and reluctance to allow suckling. Damage to other piglets may interfere with the establishment of the “pecking order” in the litter.

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin) The acquisition and absorption of adequate amounts of colostral immunoglobulins is essential to the health of ruminant, porcine, and equine neonates because they are born virtually devoid of circulating immunoglobulin. Failure of passive transfer (FPT) has been a commonly used term to describe the transfer of passive immunity (immunoglobulins, specifically IgG1 in colostrum) from the dam to the neonate. The process by which colostral immunoglobulin is absorbed is far from passive; it is an active and focused activity. Accordingly, FPT provides an incorrect summary of this process, and failure of transfer of passive immunity (FTPI) provides a more accurate descriptive term. Adequate antibody transfer is the cornerstone of all neonatal preventive health programs, but FTPI remains an important problem particularly affecting the dairy industry. Educational campaigns targeting dairy producers have been launched in the past decades, and, encouragingly, the prevalence of FTPI in dairy heifers in the United States decreased from over 40% in 1992 to 19% in 2007.1 Much of the description that follows refers to the calf because more studies on transfer of passive immunity have been conducted in calves. However, most of the information is applicable to the other species; where there are differences, these are mentioned.

NORMAL TRANSFER OF IMMUNOGLOBULINS

Immunoglobulins in colostrum are present in different concentrations. The major immunoglobulin in colostrum is IgG. IgG consists of two fractions, IgG1 and IgG2, which contribute 80% and 5% to 10%,

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)

VetBooks.ir

respectively, to the total colostral immunoglobulin concentration. IgM and IgA each account for approximately 5% of the colostral immunoglobulin content. IgG is concentrated in colostrum by an active, selective, receptor-mediated transfer from the blood of the dam across the mammary secretory epithelium. This transfer to colostrum begins approximately 4 to 6 weeks before parturition and results in colostral IgG concentrations in first milking colostrum that are several-fold higher than maternal serum concentrations. This active IgG transfer ceases suddenly at the onset of lactation, presumably in response to increased prolactin secretion around parturition.2 IgA and IgM are largely derived from local synthesis by the mammary gland rather transfer from plasma. Following ingestion by the newborn, a significant proportion of these immunoglobulins is transferred across the epithelial cells of the small intestine during the first few hours of life and transported via the lymphatic system to the blood. Immunoglobulins in blood are further varyingly distributed to extravascular fluids and to body secretions depending on the immunoglobulin class. These absorbed immunoglobulins protect against systemic invasion by microorganisms and septicemic disease during the neonatal period. Unabsorbed immunoglobulins and immunoglobulins resecreted into the gut play an important role in protection against intestinal disease for several weeks following birth. FTPI has unequivocally been associated with increased morbidity and mortality and reduced growth rates of neonates. Adequate immunoglobulin supply at birth is associated with higher firstand second-lactation milk production and decreased risk of culling during the first lactation.5 In foals, FTPI presents a significant risk for the development of illness during the first 3 months of life. Lactogenic Immunity The IgG concentration in milk falls rapidly following parturition in all species, and immunoglobulin concentrations in milk are low (Table 19-3). In the sow, the

concentration of IgA falls only slightly during the same period, and it becomes a major immunoglobulin of sows’ milk. IgA is synthesized by the mammary gland of the sow throughout lactation and serves as an important defense mechanism against enteric disease in the nursing piglet. IgA in milk is an important mucosal defense mechanism in piglets, whereas in the calf there is little IgA in milk, but some enteric protection is provided by colostral and milk IgG and IgG derived from serum that is resecreted into the intestine.

FAILURE OF TRANSFER OF PASSIVE IMMUNITY

FTPI is the major determinant of septicemic disease in most species. It also modulates the occurrence of mortality and severity of enteric and respiratory disease in early life and performance at later ages. In terms of the modulation of disease, there can be no set cut-point for circulating immunoglobulins because this cut-point will vary according to the farm, its environment, infection pressure, and the type of disease. Values are given as guidelines. FTPI in calves has been defined as a serum IgG concentration below 1000 mg/dL (10 mg/mL) when measured between 24 hours and 7 days of age. With foals, the equivalent IgG cutoff concentrations for FTPI and partial FTPI are given as 400 mg/dL and 800 mg/dL, respectively. Although a serum IgG concentration above 400 mg/dL might be adequate for healthy foals kept in a clean environment with minimal pathogen exposure, a con­ centration above 800 mg/dL is considered optimal.6 For New World camelid crias, a cut-point value for the serum IgG concentration of 1000 mg/dL measured at around 36 hours of life has been recommended.7 Rates of FTPI in dairy calves can vary widely between farms, but they were estimated to be in the range of 20% in a recent nationwide survey conducted in the United States.1 In beef calves FTPI rates tend to be lower; a recent Canadian study reported the incidence of FTPI, defined as serum IgG concentrations below 800 mg/dL, of 6% and a rate of marginal transfer of passive immunity (800 mg/dL < IgG < 1600 mg/dL) of

1849

10%.8 Failure rates in foals reported in the literature are approximate 13% to 16%. Rates in lambs are also comparatively low, and the incidence of FTPI in crias has been estimated to be around 10%.7 In animals that are fed colostrum artificially, risk for FTPI is primarily dependent on the amount or mass of immunoglobulin present in a feeding of colostrum, the time after birth that this is fed, the efficiency of its absorption from the digestive tract, and possibly also the degree of bacterial contamination.1 The mass of immunoglobulins fed is determined by the concentration of immunoglobulin in the colostrum and the volume that is fed. Feeding trials with calves suggest that a mass of at least 150  g of IgG is required in colostrum fed to a 45-kg calf to obtain adequate (≥1000  mg/dL IgG) colostral immunoglobulin concentrations in serum. In animals that suck colostrum naturally, such as foals, risk for FTPI is primarily dependent on the concentration of immunoglobulin in the colostrum, the amount that is ingested, and the time of first suckling. Inadequate colostral immunoglobulin concentration and delay in ingestion of colostrum are the two important factors in FTPI in foals.

DETERMINANTS OF TRANSFER OF COLOSTRAL IMMUNOGLOBULINS 1. Amount of immunoglobulin in colostrum fed: a. Volume of colostrum fed b. Concentration of immunoglobulins in colostrum 2. Amount of colostrum actually suckled or fed 3. Rate of abomasal or gastric emptying after colostrum ingestion 4. Efficiency of absorption of immunoglobulins by neonate 5. Time after birth of suckling or feeding 6. Time of collection of colostrum after calving (with artificial colostrum feeding) 7. Degree of bacterial contamination of colostrum

Table 19-3  Failure of transfer of passive immunity;1 concentrations and relative percentage of immunoglobulins in serum and mammary secretions of cattle and pigs CONCENTRATION (mg/ml)

Animal

Immunoglobulin

Cow

IgG1 IgG2 IgM IgA IgG IgM IgA

Sow

TOTAL IMMUNOGLOBULIN (%)

Serum

Colostrum

Milk

Serum

Colostrum

Milk

11.0 7.9 2.6 0.5 21.5 1.1 1.8

75.0 2.9 4.9 4.4 58.7 3.2 10.7

0.59 0.02 0.05 0.14 3.0 0.3 7.7

50 36 12 2 89 4 7

81 5 7 7 80 6 14

73 2.5 6.5 18 29 1 70

1850

Chapter 19 

■ 

Perinatal Diseases

VetBooks.ir

Determinants of Immunoglobulin Concentration in Colostrum Nominal concentrations of immunoglobulin in the first milking colostrum of cows and sows are shown in Table 19-4.1 Current conventional wisdom posits that high-quality bovine colostrum should contain at least 50 g/L IgG,2 and that 3 L of high-quality colostrum should be fed as soon as possible after birth.3,4 This strategy will provide the needed 150 g of colostral IgG. There can be substantial variation in the concentration of immunoglobulin in colostrum in all species, and the ingestion of a “normal” amount of colostrum that has low immunoglobulin concentration may provide an insufficient amount of immunoglobulin for protection. In a study of over 900 first-milkings colostrum from Holstein Friesian cows, only 29% of the colostrum samples contained a sufficiently high concentration of immunoglobulin to provide 100 g IgG in a 2-L volume. The equivalent percentages for 3and 4-L volume feedings were 71% and 87%, respectively. It is apparent that variation in colostral immunoglobulin concentration can be a cause of FTPI. Some causes of this variation are the following: • The concentrations of immunoglobulin in colostrum fall dramatically after parturition. The concentrations in second-milking colostrum are approximately half those in the first milking, and by the fifth postcalving milking, concentrations approach those found during the remainder of lactation. A similar situation exists with horses. The mean concentrations of IgG in colostrum of mares 3 to 28 days before foaling is greater than 1000 mg IgG/dL, whereas at parturition the mean concentrations may vary from 4000 to 9000 mg/dL. The concentrations decrease markedly to 1000 mg/dL in 8 to 19 hours after parturition. • The immunoglobulin concentration of colostrum decreases after calving even when the cow is not milked. It is important that this colostrum be milked as soon as possible after parturition. Colostrum that is collected 6 hours or later after calving has a significantly lower concentration than that collected 2 hours after calving. In a study documenting the effect of time since parturition on colostral IgG concentration, it was observed that colostral IgG concentration decreased by 3.7% during each subsequent hour after calving because of postparturient secretion of IgG-poor milk by the mammary glands. • Colostrum from cows or mares that have been premilked to reduce udder edema or from dams that leaked colostrum before parturition have low immunoglobulin concentrations, and

• •









• •

alternate colostrum should be fed for immunoglobulin transfer. In cattle, dry periods of less than 30 days may result in colostrum of lower immunoglobulin concentration. Premature foaling or the induction of premature parturition using longacting corticosteroids in cattle can result in colostrum with low immunoglobulin concentration and/or low volume. In cattle, average colostral immunoglobulin concentrations are higher in cows in third or higher lactation groups compared with younger cows. However, colostrum from all lactation numbers can produce adequate immunoglobulin mass. There is no scientific basis for not feeding first-milking colostrum from firstlactation cows. Larger-volume first-milking colostrum tends to have lower immunoglobulin concentrations than smaller-volume colostrum, presumably as a result of dilution. Immunoglobulin concentrations were found to be higher in the early temporal fractions of a single milking of firstmilking colostrum. This might suggest that segregation of the first portion of the first-milking colostrum could provide colostrum with higher immunoglobulin concentration for feeding. There are breed differences in the concentration of immunoglobulins in first-milking colostrum. In cattle, beef breeds have higher concentrations. Many dairy breeds, including Holstein Friesian, produce colostrum of relatively low immunoglobulin concentration, and a significant proportion of calves that suckle cows of these breeds ingest an inadequate mass of immunoglobulin. Channel Island breeds have a greater concentration of immunoglobulin in colostrum that Holstein Friesians. Breed differences are also seen in horses, with Arabian mares having higher colostral immunoglobulin concentrations than Standardbreds, which in turn are higher than those of Thoroughbreds. Breed differences also occur in sheep, with higher concentrations in meat and wool breeds than dairy breeds. Heat stress to cattle in the latter part of pregnancy results in lower colostral immunoglobulin concentrations. Colostral volume but not colostral immunoglobulin concentration is reduced in mastitic quarters, and it is unlikely that mastitis is a major determinant of the high rate of FTPI in dairy calves. Colostrum from cows with clinical mastitis should nonetheless not be fed because it may contain pathogens in large amounts and has unphysiologic composition.

• The pooling of colostrum in theory could avoid the variation in immunoglobulin concentration of individually fed colostrum and could provide a colostrum that reflects the antigenic experience of several cattle. In practice, colostrum pools from Holsteins invariably have low immunoglobulin concentrations because high-volume, low-concentration colostrum dilutes the concentration of the other samples in the pool. If pools are used, the diluting influence of low-immunoglobulin-concentration, high-volume colostrum should be limited by restricting any individual cow’s contribution to the pool to 9 kg (20 lb) or less. However, pooling increases the risk of disease transmission because multiple cows are represented in a pool and the pool is fed to multiple calves. This can be important in the control of Johne’s disease, bovine leukosis, Mycoplasma bovis, E. coli, and Salmonella spp. • Bacterial contamination of colostrum can have a negative effect on transfer of passive immunity. The current recommendation is that fresh colostrum should contain less than 100,000 cfu/mL total bacteria count and less than 10,000 cfu total coliform count.2 One study found that 85% of colostrums sampled from 40 farms in the United States exceeded this threshold. Colostrum that is to be fed or stored should be collected with appropriate preparation and sanitation of the cow and of the milking equipment used on fresh cows. • Pasteurization of colostrum either at 63° C (145° F) for 30 min or 72° C (162° F) for 15 s was shown to reduce colostrum IgG concentration by at least 30% and to thicken or congeal the colostrum. In contrast, pasteurization at 60° C (140° F) for 60 min was found to affect neither colostral IgG concentration nor fluid characteristics while eliminating or at least significantly decreasing the content of major pathogens, including Mycobacterium avium subps. paratuberculosis, M. bovis, E. coli, and Salmonella spp. • Old mares (older than 15 years) may have poor colostral immunoglobulin concentration. Volume of Colostrum Ingested Dairy Cows The volume of colostrum that is fed has a direct influence on the mass of immunoglobulin ingested at first feeding. The average volume of colostrum ingested by nursing Holstein Friesian calves in the first 24 hours of life is reported as 2.4 L, but there is wide variation around this mean. In natural suckling situations, calves may fail to ingest

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)

VetBooks.ir

adequate colostrum volumes before onset of the closure process and therefore absorb insufficient colostral immunoglobulin. Early assisted suckling may help avoid this. In dairy calves the volume of colostrum that is ingested can be controlled in artificial feeding systems using nipple bottle feeders or esophageal tube feeders. Bucket feeding of colostrum is not recommended because training to feed from a bucket can be associated with erratic intakes. The traditional recommendation for the volume of colostrum to feed at first feeding to calves is 2 L (2 quarts). However, only a small proportion of first-milking colostrum from Holsteins contains a sufficiently high concentration of immunoglobulin to provide 100 g IgG in a 2-L volume, and higher volumes of colostrum are required to achieve this mass intake. Some calves fed with a nipple bottle will drink volumes greater than 2 L, but others will refuse to ingest even 2 L of colostrum in a reasonable period of time, and calf rearers may lack the time or patience to persist with nipple bottle feeding until the required volume has been ingested by all calves. Larger volumes of colostrum can be fed by an esophageal feeder, and single feedings of large volumes of colostrum (3.5 to 4.0 L per 45 kg of body weight) result in the lowest percentage of calves with FTPI by allowing calves fed colostrum with relatively low immunoglobulin concentrations to receive an adequate immunoglobulin mass before closure. Feeding this volume by an esophageal feeder causes no apparent discomfort to a minimally restrained calf and was not found to negatively affect intestinal IgG absorption compared with voluntary intake of the same (large) volume.10 There is nonetheless some debate around the recommendation to systematically tube feed neonatal calves because of animal welfare concerns. In several European countries animal welfare legislation prohibits force-feeding of animals without medical indication.20 Beef Cows With beef breeds very effective colostral immunoglobulin transfer is achieved with natural sucking. This is thought to be a result of the greater vigor at birth exhibited by these calves and the higher immunoglobulin concentrations in beef colostrum, requiring a smaller volume intake to acquire an adequate mass. Natural sucking will give an adequate volume intake, and there is no need to artificially feed colostrum unless the dam is observed to refuse nursing or the calf ’s viability and sucking drive are compromised. The yield of colostrum and colostral immunoglobulins in beef cows can vary widely, and range beef heifers may produce critically low volumes of colostrum. Differences in yield can be attributed to breed or to nutritional status, although undernutrition is not an effect unless it is very severe.

Ewes Colostrum yield is high in ewes in good condition at lambing, but it may be low in ewes with condition scores of 1.5 to 2.0. Sows In sows there is also very effective colostral immunoglobulin transfer with natural sucking, and piglets average an intake of 5% to 7% of body weight in the first hour of life. There is between-sow variation in the amount of colostrum, and there can be a large variation in colostrum supply from teat to teat, which may explain variable health and performance. During farrowing and for a short period following, colostrum is available freely from the udder, but thereafter it is released in ejections during mass suckling. A strong coordinated sucking stimulus is required by the piglet for maximum release of colostrum, and this requires that the ambient temperature and other environmental factors be conducive to the optimum vigor of the piglets. Smallbirth-weight piglets, late-birth-order piglets, and piglets sucking posterior teats obtain less colostrum. All Species In all species a low-volume intake may also occur because of the following factors: • Poor mothering behavior, which may prevent the newborn from sucking; occurrence of disease; or milk fever • Poor udder and/or teat conformation so that the newborn cannot suck normally or teat seeking is more prolonged. Udder-to-floor distance is most critical, and low-slung udders can account for significant delays in intake. Bottle-shaped teats (35-mm diameter) also significantly reduce intake. • Delayed and inadequate colostral intake frequently accompanies perinatal asphyxia or acidemia because of the greatly decreased vigor of the calf in the first few hours of life. Perinatal asphyxia can occur in any breed and is greatly increased by matings resulting in fetal–maternal disproportion and dystocia. • The newborn may be weak, traumatized, or unable to suck for other reasons; a weak sucking drive can be a result of congenital iodine deficiency, cold stress, or other factors. • Disease of the periparturient dam, such as clinical hypocalcemia in cattle or the mastitis metritis agalactia complex in sows, may preclude adequate colostrum intake by offspring. • Failure to allow newborn animals to ingest colostrum may occur under some management systems. Efficiency of Absorption After ingestion of colostrum by the newborn, colostral immunoglobulins are absorbed

1851

from the small intestine, by a process of pinocytosis, into the columnar cells of the epithelium. In the newborn calf this is a very rapid process, and immunoglobulin can be detected in the thoracic duct lymph within 80 to 120 minutes of its being introduced into the duodenum. The period of absorption varies between species and with immunoglobulin class. The mechanism by which absorption ceases is not well understood, but it may be related to replacement of the fetal enterocyte. The region of maximum absorption is in the lower small intestine, and peak serum concentrations are reached by 12 to 24 hours in all species. Absorption is not limited to immunoglobulin, and proteinuria during the first 24 hours of life is associated with the renal excretion of low-molecular-weight proteins such as β-lactoglobulin. Feeding Methods, “Closure of the Gut,” and Immunoglobulin Absorption Under normal conditions complete loss of the ability to absorb immunoglobulin (closure of the gut) occurs by 24 to 36 hours after birth in all species, and there is a significant reduction in absorptive ability (as much as 50% in some studies but minimal in others) by 8 to 12 hours following birth. The time from birth to feeding is a crucial factor affecting the absorption of colostral immunoglobulin in all species, and any delay beyond the first few hours of life, particularly after 8 hours, significantly reduces the amount of immunoglobulin absorbed. The recommendation is that all neonates should be fed colostrum within the first 2 hours of life. Natural Sucking Natural sucking is the desired method of intake of colostrum and is the most efficient, but it is influenced by the sucking drive and vigor of the neonate at birth. Newborns that suck colostrum can achieve very high concentrations of colostral immunoglobulin, and the efficiency of absorption is best with this feeding method. However, in dairy calves natural sucking is commonly associated with a high rate of FTPI because of delays in sucking coupled with low intake. Rates of FTPI in calves allowed to obtain colostrum via voluntary nursing reported in the literature can be as high as 40% to 60%.1 Many factors influence the occurrence of delayed sucking, but calf vigor and birthrelated asphyxia are the most important. Parity of the dam, conformation of the udder, and breed were also found to be significantly associated with the rate of FTPI. One older study reported that 46% of all calves born to multiparous cows had failed to nurse within 6 hours of birth compared with 11% of calves of primiparous cows.11 Jersey calves have better rates of successful transfer of passive immunity with natural sucking than do Holsteins Friesians.

1852

Chapter 19 

■ 

Perinatal Diseases

VetBooks.ir

Artificial Feeding In contrast, when calves are fed colostrum artificially, minimal delays from birth to the time of colostrum feeding occur, and maximal colostrum immunoglobulin absorption results. In breeds such as Holstein Friesians, where colostral immunoglobulin concentrations tend to be low and maximal efficiency of absorption is necessary, the logical way to minimize risk of FTPI is to feed the maximum volume of colostrum that is well tolerated within the first few hours of life. The published literature consistently reports higher calf serum IgG concentrations and a lower rate of FTPI in response to larger colostrum feeding volumes.2,10,12 Other Influences Even with the best available on-farm colostrum-selection methods, large colostrumfeeding volumes are essential to minimize the risk of FTPI in breeds with relatively low colostral immunoglobulin concentrations. The method is particularly advantageous where time constraints of other farm activities limit the time available for calf feeding. The major detrimental influence on absorptive efficiency of immunoglobulin is delayed feeding after birth. Other factors that may affect absorptive efficiency include the following: • Perinatal asphyxia or acidemia may have both direct and indirect effects on colostral immunoglobulin transfer. Asphyxia has a major effect on subsequent sucking drive, and acidemic calves ingest far less colostrum than calves with more normal acid–base status at birth. In carefully controlled colostrum feeding studies, a significant negative correlation between the degree of hypercapnia and the efficiency of absorption of colostral immunoglobulin in the first hours of life was demonstrated. However, this effect was only transient because there was no difference in serum IgG concentration at the time of gut closure between normoxic and hypoxic calves. • In one early study, a mothering effect was reported in which calves remaining with their dams absorbed colostral immunoglobulin more efficiently than calves removed immediately to individual pens. However, other studies have shown much smaller or no effects of mothering using similar experimental designs. The different results of these studies have not been reconciled. • There can be seasonal and geographic variations in transfer of immunoglobulin in calves, although these are not always present on farms in the same area, and their cause is unknown. Where seasonal variation occurs in temperate climates, the mean monthly serum IgG concentrations are lowest in the winter and increase during

the spring and early summer to reach their peak in September, after which they decrease. The cause is not known, but a decrease in sucking drive is observed in colder months and may contribute. In subtropical climates, peak levels occur in the winter months, and low levels are associated with elevated temperatures during the summer months. Heat stress in late pregnancy will reduce colostral immunoglobulin concentration, but high ambient temperature is a strong depressant of absorption, and the provision of shade will help to obviate the problem. • The efficiency of absorption may be decreased in premature calves that are born following induced parturition using long-acting corticosteroids; in contrast, medical induction of parturition with short-acting corticosteroids in cattle does not interfere with the efficiency of absorption of immunoglobulins in calves. • The absorption of small volumes (1 to 2 L) fed by an esophageal feeder is usually suboptimal and inferior to the absorption after sucking the same small volume.10 This effect may at least in part be attributable to retention of some colostrum in the immature forestomaches for several hours. The calf will feel satiated and not inclined to suck naturally for the next few hours. • A trypsin inhibitor in colostrum may serve to protect colostral IgG from intestinal degradation. It varies in concentration between colostrums. The addition of a trypsin inhibitor to colostrum improves immunoglobulin absorption. • In a study of mare-associated determinants of FTPI in foals (based on serum Ig measurements), there was a trend to increase rates of FTPI in foals from mares aged over 12 years, but no significant association with age, parity, or gestational age of foals over 325 days was found. There was an association with season, with a lower incidence in the late spring compared with foals born earlier in the year and with a foal score based on a veterinary score of foal health and “fitness.” Traditionally it has been considered that the movement of animals, either the dam just before parturition or the newborn animal during the first few days of life, is a special hazard for the health of the calf. The postulated reason is that the dam may not have been exposed to pathogens present in the new environment and thus not have circulating antibodies against these pathogens. The newborn animal may be in the same position with regard to both deficiency of antibodies and exposure to new infections. Although this may be the case in some

situations, the developing practice of contract-rearing of dairy heifers away from the farm to be brought back as close-up springers and the practice of purchase of close-up heifers on the farm are not associated with appreciable increase in mortality in their calves. Decline of Passive Immunity Colostral antibody concentrations in blood fall quickly after birth and have usually disappeared by 6 months of age. In the foal, they have fallen to less than 50% of peak level by 1 month of age and to a minimum level between 30 and 60 days. This is the point at which naturally immunodeficient foals are highly susceptible to fatal infection. In calves, the level of IgG declines slowly and reaches minimum values by 60 days, in contrast to IgM and IgA, which decline more rapidly and reach minimum values by approximately 21 days of age. The half-lives for IgG, IgM, and IgA in calves are approximately 20, 4, and 2 days, respectively, and the half-lives of IgGa, IgGb, IgG(T), and IgA in foals are approximately 18, 32, 21, and 3.5 days, respectively. Immunologic competence is present at birth, but endogenous antibody production does not usually reach protective levels until 1 month, and maximum levels are not reached until 2 to 3 months of age. The endogenous production of intestinal IgA in the piglet begins at about 2 weeks of age and does not reach significant levels until 5 weeks of age. Foals that acquire low concentrations of immunoglobulins from colostrum may experience a transitory hypogammaglobulinemia at several weeks of age as the levels fall and before autogenous antibodies develop. They are, as expected, more subject to infection than normal.

OTHER BENEFITS OF COLOSTRUM

In addition to its immunoglobulin content, colostrum contains considerably more protein, fat, vitamins, and minerals than milk and is especially important in the transfer of fat-soluble vitamins. It has anabolic effects, and lambs that ingest colostrum have a higher summit metabolism than colostrum-deprived lambs. Colostrum also contains growth-promoting factors that stimulate DNA synthesis and cell division, including high concentrations of insulin-like growth factor (IGF)-1. Colostrum contains approximately 1 × 106 leukocytes/mL, and several hundred million are ingested with the first feeding of colostrum. In calves, 20% to 30% of these are lymphocytes and cross the intestine into the circulation of the calf. It is postulated that they have importance in the development of neonatal resistance to disease, but there is little tangible evidence. Calves fed colostrum depleted of leukocytes are thought to be more poorly protected against neonatal disease than those fed normal colostrum.

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)

VetBooks.ir

ASSESSMENT OF TRANSFER OF PASSIVE IMMUNITY Because of the importance of transfer of colostral antibodies to the health of the neonate, it is common to quantitatively estimate the levels of immunoglobulins, or their surrogates, in colostrum and in serum to predict risk of disease and to take preventive measures in the individual or to make corrective management changes where groups of animals are at risk.

within 20 min, it only provides a pass/fail result using a cutoff value of 10 mg/mL.2 Turbidimetric Immunoassay The turbidimetric immunoassay (TIM) is commercially available and can be run on a handheld chemistry analyzer to be used with bovine serum. In a preliminary study conducted at the University of Minnesota, the test was found to be more accurate than indirect tests such as serum refractometry.

Assessment in the Individual Animal When samples are taken from an individual animal to determine the risk for infection, sampling is undertaken early so that replacement therapy can be given promptly if there has been inadequate transfer. IgG is detectable in serum 2 hours following a colostrum feeding and sampling at 8 to 12 hours after birth will give a good indication of whether early sucking has occurred and has been effective in transfer. This type of monitoring is commonly performed in foals and camelid crias.7,13 There are a number of different tests that can be used; some are quantitative and others semiquantitative. In calves, sampling may be undertaken for similar reasons, but the cost of replacement therapy is limiting.

Zinc Sulfate Turbidity Test The zinc sulfate turbidity test is based on a selective precipitation reaction of the salt with high molecular weight proteins such as immunoglobulin (not specifically IgG). The test is commonly used with a test solution containing 200 mg/L zinc sulfate but was found to have poor specificity and would only classify 69% of tested calves correctly. Increasing the zinc sulfate con­ centration from 200 to 350 mg/L considerably improved the specificity and positive predictive values of the test, but this test modification is not widely used.15 Another inconvenience is that hemolyzed blood samples will give artificially high readings, and the reagent must be kept free of dissolved carbon dioxide.

Assessment Tests on Serum Sampling to monitor the efficacy of a farm policy for feeding and handling colostrum, to evaluate the passive immunity status in calves to be purchased, or to determine the rates of FTPI in investigations of neonatal disease can be conducted at any time in the first week of life after 24 hours with most tests. Numerous tests are currently available, some of which directly measure serum IgG concentration and some of which estimate the IgG concentration based on the serum concentration of the total globulin or other protein fractions.

Sodium Sulfite Precipitation Test The sodium sulfite precipitation test is based on the selective precipitation of highmolecular-weight proteins with sodium sulfate at different concentrations. Test solutions of 14%, 16%, and 18% sodium sulfite are commonly used, and the development of turbidity at a certain concentration allows for a crude estimate of the serum immunoglobulin concentration; the lower the concentration at which turbidity occurs, the higher is the concentration of immunoglobulin. Particularly the use of the 14% and 16% sodium sulfite solutions was found to result in an unacceptably high percentage of calves being misclassified as FTPI while having adequate serum immunoglobulin concentrations.15

Radial Immunodiffusion The radial immunodiffusion (RID) is based on the precipitation of antigen and antibody to an insoluble precipitin complex and thus directly measures IgG concentration in serum or plasma. The RID is considered the reference method to measure serum/plasma IgG, but it takes at least 24 h to perform and thus longer than is desirable for most clinical purposes. In a recent study two commercial RID test kits for calves were compared, and a large bias and wide limits of agreements between the two tests were found, which has raised questions about the reliability of the results.21 Lateral-Flow Immunoassay The lateral-flow immunoassay is a calf-side test directly measuring IgG in serum or plasma with reportedly high sensitivity and specificity. Although the test can be performed on-site and results are available

Serum γ-Glutamyltransferase Activity Serum γ-glutamyltransferase (GGT) activity has been used as a surrogate for determining the efficacy of transfer of passive immunity in calves and lambs (not in foals). GGT activity is high in the colostrum of ruminants (but not horses), and serum GGT activity in calves and lambs that have sucked or been fed colostrum is 60 to 160 times greater than normal adult serum activity and correlates moderately well with serum IgG concentrations. The half-life of GGT from colostrum is short, and serum GGT activity falls significantly in the first week of life. Serum GGT values equivalent to a serum IgG concentration of 10  mg/mL are 200  IU/L on day 1 of life and 100  IU/L on day 4. Serum GGT concentrations less than 50  IU/L indicate FTPI.

1853

Serum Total Protein Measuring total protein concentrations in serum or plasma with a refractometer is a practical, rapid, and inexpensive method to estimate the immunoglobulin concentration by extrapolating it from the total protein concentration. Despite the indirect nature of the test, there is a reliable correlation between the refractometer reading and total immunoglobulin concentration measured by RID. In healthy calves a serum total protein of 5.5 g/dL or greater is associated with adequate transfer of passive immunity. Serum total protein has a good predictive value for fate of the newborn, and the facile and practical nature of the test and its predictive ability commend it for survey studies in calves and lambs but not foals. Cut-points will vary with the environment and the infection pressure to the calves. The sensitivity of the test is maximal using a cut-point of 5.5 g/dL, and the specificity is maximal at a cut-point of 5.0 g/dL. Because serum total protein concentration measured by refractometry can result in an incidental misclassification of an individual calf, this test is primarily recommended as a screening tool to assess the colostrum management on a herd level, but not as diagnostic tool for an individual animal. Herd screening could be conducted by testing a minimum of 12 calves on a farm between 24 hours and 7 days old. At least 80% of tested calves should have serum protein concentrations above 5.5 g/dL to consider the colostrum management satisfactory at the herd level. Serum total protein concentration can also be estimated using the same Brix refractometer used for measuring colostral IgG concentration, with an appropriate adjustment factor.14 Glutaraldehyde Coagulation Test The glutaraldehyde test was initially introduced to identify hypergammaglobulinemia in adult cattle with chronic inflammatory disease. The semiquantitative test is based on a clotting reaction of glutaraldehyde in the presence of high immunoglobulin concentration, where the time to clot formation is negatively correlated with the serum IgG.16 A modified glutaraldehyde coagulation test is also available for the detection of hypogammaglobulinemia in neonatal calves, but it is less accurate.15 The test may yield false-positive results with hemolysis and is difficult to quantitate. Latex Agglutination Test A commercial latex agglutination test is available for horses. It is rapid and provides semiquantitative results, but results were reported to be inconsistent. ELISA Snap-Test ELISA snap-tests are foal-side immunologic tests directly measuring IgG in a

1854

Chapter 19 

■ 

Perinatal Diseases

VetBooks.ir

semiquantitative manner. Test kits are commercially available for foals and have been available for calves. In foals the available snap-tests were found to be rapid and accurate. Monitoring Colostrum Brix Refractometry The most accurate and practical way to ensure that an adequate colostral mass is fed is to test the colostrum using a Brix refractometer (the digital version is preferred). This instrument was designed for use in food processing but was adapted in the late 1970s to provide a low-cost test of colostrum quality. A Brix refractometer value of 21% or 22% or higher indicates acceptable colostrum (same value for fresh or frozen samples; approximately equivalent to a colostral IgG concentration of 50 g/L); colostrum with a value below 21% or 22% should be discarded.17,18 Specific Gravity Specific gravity, determined by refractometry, can be used as a measure of the immunoglobulin content of colostrum. In mares the concentration of immunoglobulin in colostrum is highly correlated with the specific gravity of the colostrum, which in turn is highly correlated with the serum immunoglobulin levels achieved in foals. Temperature-corrected measurements are most accurate. Measurement of colostrum specific gravity provides a rapid and easy method of identifying foals likely to be at a high risk for FTPI and the need to provide them with colostrum of a higher Ig content. To prevent FTPI, it is recommended that the colostral specific gravity should be equal to or greater than 1.060, and the colostral IgG concentration should be a minimum of 3000  mg/dL. In cattle the relation of specific gravity of colostrum to colostral immunoglobulin concentration is linear but is better in Holstein Friesian than in Jersey cows. The measurement is simple, but there is a correction for temperature, and air trapped in colostrum taken by a milking machine can give a false reading if the measurement is taken too quickly after milking. The cutpoint recommended to distinguish moderate from excellent colostrum has been set at 1.050, approximating an IgG concentration of 50 g/L, and is based on the amount of immunoglobulin required for a 2-L (2-quart) feeding. Specific gravity is not a perfect surrogate for immunoglobulin concentration with cattle colostrum. It has good negative prediction, but it will falsely pass 2 out of 3 colostrums that have unacceptably low immunoglobulin concentrations. An analysis of first-milking colostrum in midwestern U.S. dairies found that specific gravity differed among breeds and was influenced by month of calving, year of calving, lactation number, and protein yield in

previous lactation and that it was more closely associated with colostrum protein concentration (r = 0.76) than IgG1 concentration (r = 0.53).

Colostrum Colostrum can be stripped from the dam and fed fresh, or the neonate can be fed stored (banked) colostrum.

Glutaraldehyde Test This test for mare colostrum is available commercially and is reported to have a high predictive value for colostrums that contain more than 38 mg/mL of IgG and have a specific gravity greater than 1.060.

Colostrum for Banking With dairy cows, first-milking colostrum from a cow with a first-milking yield of less than 10 kg should be used. The temptation for the farmer is to store the leftover from the feeding of large-volume colostrum. The leftover colostrum should not be used because it has a high probability of containing a low immunoglobulin concentration. Colostrum from mares should have a specific gravity of 1.060 or more, and 200 mL can be milked from a mare before the foal begins sucking.

ELISA Recently a cow-side immunoassay kit has become available commercially in the United States. The kit provides a positive or a negative response, with the cut-point being a concentration of 50g/L of IgG in colostrum, and has accuracy sufficient to recommend its use for rejection of colostrums with low immunoglobulin concentration.

CORRECTION OF FAILURE OF TRANSFER OF PASSIVE IMMUNITY Oral Therapy Oral therapy can be considered in individual animals (generally foals and crias), provided that FTPI—or the risk thereof—is diagnosed and the treatment is administered before gut closure (i.e., not later than 18 h of life). For foals, oral administration of at least 0.5 L frozen equine colostrum of good quality (specific gravity > 1.060) that has been properly stored and thawed is recommended. Alternatives include colostrum substitutes containing lyophilized IgG or good-quality bovine colostrum. The latter option is probably the least effective and requires at least 4 L of good-quality (specific gravity >1.050) colostrum. Parenteral Immunoglobulins Blood transfusion is commonly used in food animal practice, and the method is described elsewhere in this text. Fresh plasma from a random donor or purified hyperimmune plasma that is commercially available for foals and crias in some countries are alternatives. Large amounts are required to obtain the required high serum concentrations of immunoglobulins, and intravenous infusion can be accompanied by transfusion-type reactions.

AVOIDANCE OF FAILURE OF TRANSFER OF PASSIVE IMMUNITY

With all species, with the exception of dairy calves, the common practice is to allow the newborn to suck naturally. The policy for avoidance of FTPI with naturally sucking herds should be to provide supplemental colostrum by artificial feeding of those neonates with a high risk for FTPI, based on the risk factors detailed earlier. In the dairy calf, rates of FTPI with natural sucking are so high that many farms opt to remove the calf at birth and feed colostrum by hand to ensure adequate intake.

Storage of Colostrum Colostrum can be kept at refrigerator temperature for approximately 1 week without significant deterioration in immunoglobulins; bacteria counts, however, may reach unacceptably high levels (above 100,000 cfu/ mL) after 2 days in refrigerated milk.2 Addition of potassium sorbate in a 0.5% final solution impairs bacterial growth for several days.2 The addition of 5 g of propionic or lactic acid per liter extends the storage life to 6 weeks, but, more commonly, colostrum is frozen for storage. Frozen colostrum, at −20° C (−4° F), can be stored for at least 1 year, and there is no impairment in the subsequent absorption of immunoglobulins. Frozen colostrum should be stored in flat plastic bags in the amount required for a feeding, which facilitates thawing. Thawing should be at temperatures below 55° C (131° F). Higher temperatures and microwave thawing result in the deterioration of immunoglobulins and antibodies in frozen colostrum and frozen plasma. Pasteurization of Colostrum There are several indications for pasteurization of colostrum. This procedure can be a suitable instrument in a program for the control of specific infectious diseases, such as paratuberculosis, salmonellosis, or M. bovis infection, but it can also be useful to ameliorate calf health by improving colostrum quality and reducing the exposure of the neonate to pathogens. On-farm pasteurization of bovine colostrum for 60 min at 60° C (140° F) results in elimination or at least significant reduction of bacterial contamination without impairing fluid characteristics or availability of IgG for intestinal absorption.9 One recent study reported significantly higher serum IgG concentrations at 24 h of life when calves were fed pasteurized colostrum compared with calves receiving the same quality and amount of raw colostrum.19 The authors attributed this effect to reduced bacterial interference with intestinal IgG absorption. Pasteurization extends the shelf life of refrigerated colostrum without

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)

VetBooks.ir

additives to 8 to 10 days when stored in clean, sealed containers. Cross-Species Colostrum Colostrum from another species can be used to provide immunologic protection when same-species colostrum is not available. Bovine colostrum can be fed to a number of different species. Although absorption of immunoglobulin occurs and significant protection can be achieved, the use of crossspecies colostrum is not without risk, and the absorbed immunoglobulin has a short halflife. Bovine colostrum has been successfully used for many years to improve the survival rate of hysterectomy-produced artificially reared pigs. It has also been used as an alternate source of colostral antibody for rearing goats free of caprine arthritis–encephalitis (CAE). Colostrum from some cows can result in the development of hemolytic anemia, occurring at around 5 to 12 days of age, in lambs and kids because the IgG of some cows attaches to the red cells and their precursors in bone marrow, resulting in red cell destruction by the reticuloendothelial system. Bovine colostrum can be tested for “antisheep” factors by a gel precipitation test on colostral whey, but this test is not generally available. Bovine colostrum can provide some protection to newborn foals against neonatal infections, and protection appears to result from factors in addition to the immunoglobulins, which have a short halflife in foals. Colostrum Supplements In recent years there has been a move to develop supplements or even replacements for colostrum to feed calves. These have been attempted using IgG concentrated from bovine colostrum, milk whey, eggs, or bovine serum. The search for colostrum substitutes or colostrum replacers has been prompted by the problem of the variability of IgG concentration in natural colostrum. It has also been prompted by possible limitations of availability of high-quality colostrum on dairy farms as a result of discarding colostrum from cows that test positive for diseases that can transmit through colostrum, such as paratuberculosis, bovine leukosis, and M. bovis. There is evidence that the inclusion of colostrum replacer (CR) or colostrum supplement (CS) products can impair the efficiency of colostral immunoglobulin, and if they are fed, they should be fed after normal colostrum rather than mixed into the colostrum. It has been proposed that the distinction between a colostrum supplement and a colostrum replacer should be the immunoglobulin mass contained in the product, with a colostrum supplement containing less than 100 g IgG per dose and a colostrum replacer having sufficient immunoglobulin mass in a dose to result in a serum IgG concentration greater than 10 mg/mL following a feeding.

Furthermore, CR products are formulated to provide adequate protein, energy, minerals, and vitamins to completely replace colostrum, which is not the case for CS supplement products. When fed as the sole source of immunoglobulin to colostrum-deprived calves, CS products achieve circulating concentrations of immunoglobulin that are lower than those achieved by natural colostrum containing equivalent amounts of immunoglobulin. A large mass of immunoglobulin is required for acquisition of adequate serum immunoglobulin concentrations. Calves fed a colostrum replacement containing a high mass (250 g) of an IgG derived from bovine serum and fed at 1.5 and again at 13.5 hours after birth achieved equivalent serum IgG concentrations to calves fed normal colostrum and showed no difference in gain or health parameters during the first 4 weeks of life. However, the performance of commercially available products for IgG supplementation varies greatly, with many of them faring badly. The choice of a specific product should therefore be based on the availability of convincing data supporting the efficacy of the product in question. The use of colostrum replacers should be limited to situations where sufficient amounts of colostrum of adequate quality are unavailable. There can be little justification for more widespread use, particularly because there are limited independent health-related publications documenting their efficacy. Also, as mentioned earlier, in addition to immunoglobulin, natural colostrum contains various substances important to neonatal physiology. Lacteal-Secretion-Based Preparations Colostrum supplements prepared from whey or colostrum are available commercially in many countries. Depending on the manufacturer, they contain varying amounts of immunoglobulin, but significantly less than first-milking colostrum. The amount of immunoglobulin contained varies, but the recommendations for feeding that accompany these products indicate that they will supply approximately 25% or less of the immunoglobulin required to elevate calf serum IgG concentrations above 1000 mg/ dL. There is a further problem in that the immunoglobulins in products made from colostrum or whey are poorly absorbed, and trials assessing their ability to increase circulating immunoglobulins when fed with colostrum have generally shown little improvement and no improvement in health-related parameters. Bovine-Serum-Based Preparations Colostrum supplements prepared from bovine serum are also available commercially, but regulations governing the feeding of blood or blood products to calves (risk reduction for bovine spongiform

1855

encephalopathy) may limit their availability in some countries. The absorption of immunoglobulin from these bovine-serumderived commercial products appears better than from milk-protein-derived products, and consequently they are also marketed as colostrum replacers. The IgG in a commercially available bovine serum colostrum replacer has been shown to be effectively absorbed when fed to newborn lambs. The feeding of 200 g of IgG in the first 24 hours of life resulted in a mean plasma concentration of 1800 mg/dL. Administration of Colostrum Foals Foals should be allowed to suck naturally. The specific gravity of the mare’s colostrum can be checked at foaling; if this is less than 1.060, supplemental colostrum may be indicated. Foals that do not suck, or that have serum IgG concentrations less than 400 mg/ dL at 12 hours of age, or that require supplementation for other reasons, should be fed colostrum with a specific gravity of 1.060 or more at an amount of 200 mL at hourly feedings. Dairy Calves Assisted Natural Sucking Leaving the newborn dairy calf with the cow is no guarantee that the calf will obtain sufficient colostrum, and a high proportion of dairy calves fail either to suck early or to absorb sufficient immunoglobulins from ingested colostrum. This problem can be alleviated to some extent by assisted natural sucking, but this can fail because not all calves requiring assistance are detected. An alternate approach is to milk 2 L of colostrum from the dam, bottle feed each calf as soon after birth as possible, then leave the calf with the cow for 24 hours and allow it to suck voluntarily. Although this will not be as effective as a system based entirely on artificial feeding of selected colostrum, it is an approach that is suitable for the smaller dairy farm. Artificial Feeding Systems With artificial feeding systems, the calf is removed from the dam at birth and fed colostrum by hand throughout the whole absorptive period. Nipple bottle feeding can be used, with 2 L of colostrum given every 12 hours for the first 48 hours of life. The first feeding is usually milked from the cow by hand, and the remaining feedings are from the colostrum obtained from the cow after the first machine milking. With care and patience, this system can result in good transfer of passive immunity in all calves except those born to dams that have very low concentrations of immunoglobulin in their colostrum. Unfortunately, with Holstein Friesians this can be a significant percentage. An extension of this system is to bottle feed at the same frequency but to feed stored

1856

Chapter 19 

■ 

Perinatal Diseases

VetBooks.ir

colostrum selected for its superior immunoglobulin content. Bottle feeding of newborn calves requires considerable patience, and its success is very much dependent on the calf feeder and on the availability of the feeder’s time when faced with a calf that has a slow intake. Where the diligence of the calf feeders is poor, or where there is a time constraint on their availability, the feeding of a large volume of colostrum (4 L to a 45 kg calf) by esophageal feeder at the initial feeding immediately after birth can be a successful practice. The large-volume feeding also allows the delivery of an adequate mass of immunoglobulin with colostrum that has low immunoglobulin concentrations without impairing the intestinal IgG absorption rate compared with voluntary intake of the same large amount of colostrum.10 The practice usually uses stored colostrum, and the feeding can be achieved within a few minutes. It can be supplemented by bottle feeding of a second feeding at 12 hours of life. The practice of feeding stored colostrum as the sole source of colostrum is limited to larger dairy herds, but it does allow the selection of superior colostrum for feeding, with selection based on weight and specific gravity as detailed earlier. Beef Calves Beef calves should be allowed to suck naturally, and force-feeding of colostrum to beef breeds should not be practiced unless there is obvious failure of sucking. Where colostrum is required, as with weak beef calves, calves with edematous tongues, and calves that have been subjected to a difficult birth, it can be administered with an esophageal feeder or a stomach tube. Lambs Lambs are allowed to suck naturally, but there can be competition between siblings for colostrum; one large single lamb is capable of ingesting, within a short period of birth, all the available colostrum in the ewe’s udder. Lambs require a total of 180 to 210 mL colostrum/kg body weight during the first 18 hours after birth to provide sufficient energy for heat production. This amount will usually provide enough immunoglobulin for pro­ tection against infections. Supplemental feeding of colostrum may be advisable for lambs from multiple birth litters, lambs that lack vigor, and those that have not nursed by 2 hours following birth. This can be done with a nipple bottle or an esophageal feeder. Piglets Colostral supplementation is not commonly practiced with piglets. An immunoglobulin dose of 10 g/kg body weight on day 1 followed by 2 g/kg on succeeding days for 10 days is sufficient to confer passive immunity on the colostrum-deprived pig.

FURTHER READING Barrington GM, Parish SM. Bovine neonatal immunology. Vet Clin North Am Food Anim Pract. 2001;17:463-476. Black L, Francis ML, Nicholls MJ. Protecting young domestic animals from infectious disease. Vet Annu. 1985;25:46-61. Godden S. Colostrum management for dairy calves. Vet Clin North Am Food Anim Pract. 2008;24:19-39. McGuirk SM, Collins M. Managing the production, storage, and delivery of colostrum. Vet Clin North Am Food Anim Pract. 2004;20:593-603. Mellor D. Meeting colostrum needs of lambs. In Pract. 1990;12:239-244. Norcross NL. Secretion and composition of colostrum and milk. J Am Vet Med Assoc. 1982;181:1057. Quigley JD, Drewry JJ. Nutrient and immunity transfer from cow to calf pre- and postcalving. J Dairy Sci. 1998;81:2779-2790. Rooke JA, Bland IM. The acquisition of passive immunity in the newborn piglet. Livest Prod Sci. 2002;78:13-23. Staley TE, Bush LJ. Receptor mechanism of the neonatal intestine and their relationship to immunoglobulin absorption and disease. J Dairy Sci. 1985;68:184-205. Weaver DM, Tyler JW, VanMetre D, Hoetetler DE, Barrington GM. Passive transfer of colostral immunoglobulins in calves. J Vet Intern Med. 2000;14:569-577.

REFERENCES

1. Beam AL, et al. J Dairy Sci. 2009;92:3973-3980. 2. Godden S. Vet Clin North Am Food Anim Pract. 2008;24:19-39. 3. Morin DE, et al. J Am Vet Med Assoc. 2010;237:420-428. 4. Mokhber-Dezfooli MR, et al. J Dairy Sci. 2012;95:6740-6749. 5. Faber SN, et al. Prof Anim Sci. 2005;21:420-425. 6. McCue PM. Am J Vet Res. 2007;68:1005-1009. 7. Whitehead CE. Vet Clin North Am Food Anim Pract. 2009;25:353-366. 8. Waldner CL, Rosengren LB. Can Vet J. 2009;50:275-281. 9. Godden S, et al. J Dairy Sci. 2006;89:3476-3482. 10. Godden SM, et al. J Dairy Sci. 2009;92:1758-1765. 11. Edwards SA, Broom DM. Res Vet Sci. 1979;26:255-256. 12. Godden SM, et al. J Dairy Sci. 2009;92:1750-1757. 13. Austin SM. Equine Vet Educ. 2013;25:585-589. 14. Deelen SM, et al. J Dairy Sci. 2014;97:1-7. 15. Weaver DM, et al. J Vet Intern Med. 2000;14:569-577. 16. Metzner M, et al. J Vet Med A Physiol Pathol Clin Med. 2007;54:449-454. 17. Bielmann V, et al. J Dairy Sci. 2010;93:3713-3721. 18. Quigley JD, et al. J Dairy Sci. 2013;96:1148-1155. 19. Johnson J, et al. J Dairy Sci. 2007;90:5189-5198. 20. Lorenz I, et al. Ir Vet J. 2011;64:10. 21. Ameri M, Wilkerson MJ. J Vet Diagn Invest. 2008;20:333-336.

Clinical Assessment and Care of Critically Ill Newborns The following discussion focuses on care and treatment of critically ill foals, although the principles are applicable to any species. The increasing availability of secondary and

tertiary care for ill newborns has allowed the development of sophisticated care for newborns of sufficient emotional or financial value.1 This level of care, at its most intensive, requires appropriately trained individuals (both veterinarians and support staff) and dedicated facilities. True intensive care of newborns requires 24-hour monitoring. The following discussion is not a comprehensive guide to intensive care of newborns, but is rather an introduction to the general aspects of advanced primary or basic secondary care. Sophisticated interventions, such as mechanical ventilation and cardiovascular support, are mentioned but not discussed in detail.

CLINICAL EXAMINATION

Initial assessment of an ill newborn should begin with collection of a detailed history, including length of gestation, health of the dam, parturition, and behavior of the newborn after birth, including the time to stand and to commence nursing activity. Physical examination should be thorough, with particular attention to those body systems most commonly affected. A form similar to that in Figure 19-4 is useful in ensuring that all pertinent questions are addressed and that the physical examination is comprehensive. Examination of ill neonates should focus on detection of the common causes of disease in this age group: sepsis, either focal or systemic; prematurity or dysmaturity; metabolic abnormalities (such as hypoglycemia or hypothermia); birth trauma; diseases associated with hypoxia; and congenital abnormalities. Detailed descriptions of these conditions are provided elsewhere in this chapter. Sepsis Sepsis is an important cause of illness in neonates that can manifest as localized infections without apparent systemic signs, localized infections with signs of systemic illness, or systemic illness without signs of localized infection.2 Localized infections without signs of systemic illness include septic synovitis or osteomyelitis and omphalitis. Signs of these diseases are evident on examination of the area affected and include lameness, distension of the joint, and pain on palpation of the affected joint in animals with synovitis or osteomyelitis and an enlarged external umbilicus with or without purulent discharge in animals with infections of the umbilical structures. Specialized imaging and hematologic and serum biochemical examinations (see following discussion) are useful in confirming the infection. Systemic signs of sepsis include depression, failure to nurse or reduced frequency of nursing, somnolence, recumbency, fever or hypothermia, tachypnea, tachycardia, diarrhea, and colic, in addition to any signs of

Failure of Transfer of Passive Immunity (Failure of Transfer of Colostral Immunoglobulin)

1857

VetBooks.ir

Foal Examination Protocol (age< 1mon) The Ohio State University Veterinary Teaching Hospital

Special considerations:

Clinician: Student:

History

Date:

Time:

AM/PM

Mare Age:

Problems with previous foals?

No of previous foals:

No

Uterine infections/Vaginal discharge? liiness during pregnancy?

No

Milk dripping?

How long?

No No

Vaccinations?

No

Deworming?

Yes

Yes

Yes

Yes

What/When?

Yes Yes

No

When?

Feeding: Dystocia?

No

Early cord rupture?

No

Normal

Premature placental separation?

Yes

days)

Yes

No

No

Yes

Condition of placenta:

Yes

No

Meconium staining? Colostrum quality:

overdue (

early

Yes

Placenta completely passed? Udder:

on term

Duration of prenancy:

Breeding date:

Abnormal Normal

Amount:

Low-quality

Normal

Reduced

Foal Spontaneous breathing?

No

Nursing normally?

Yes

Behavior normal? Urination?

No

No No

Yes

Time to stand: Colostrum/Milk given? lgG tested?

Yes Meconium passed?

Yes

Medications given?

No

Yes

Umbilicus treated?

No

Yes

Time to nurse:

No

Yes

No

Yes

Enema given?

No

Yes

Presenting complaint:

Previous treatment: The Ohio State University Form–209046

Foal Examination Protocol

Fig. 19-4  Examples of forms used to document and record historical aspects and findings on physical examination of foals less than 1 month Continued

of age.

1858

Chapter 19 

■ 

Perinatal Diseases

VetBooks.ir

Physical Examination Temperature:

ºF

Time:

Date:

Pulse rate:

/min

Respiratory rate:

/min

AM/PM

Body weight:

kg /

lb

Inspection: Behavior: no

Signs of prematurity?

yes (

Haircoat

Forehead

Ears

Joints

Tendons

)

Skin and haircoat: Body condition: good

Suckle reflex: Eyes:

weak

moderate

Entropion (L)(R)

normal

none

Uveitis (L)(R)

Corneal ulcer (L)(R)

Cardiovascular moderate

strong

Pulse quality:

weak /

CRT:

Mucous membranes: collapsed

normal

Jugular veins:

irregular

regular

Intensity:

Cardiac auscultation: HR: Murmurs:

no

Skin turgor:

sec.

Catheter

distended Rhythm:

regular

irregular

yes

Respiration no

Nasal discharge:

Cough:

yes Auscultation:

normal:

Lymph nodes:

no

yes

normal:

Gl tract Colic:

no

yes

Abd. distention:

GL sounds:

no

Digital palpation/Meconium:

Fecal consistency: Urogenital Umbilicus: Urination:

normal no

yes

straining

Scrotum/Testes – Vulva/Vagina:

Musculoskeletal Joints:

normal

Lameness:

no

yes

Deformations/Angular limb deformities:

no

yes

Neurologic: normal Seizures:

no

Senior Student:

yes

Attending Clinician:

Foal Examination Protocol Fig. 19-4, cont’d

normal

yes

left

right

Clinical Assessment and Care of Critically Ill Newborns

VetBooks.ir

localized disease. Fever is a specific, but not sensitive, sign of sepsis in foals. The presence of petechia in oral, nasal, ocular, or vaginal mucous membranes, the pinna, or coronary bands is considered a specific indicator of sepsis, although this has not been documented by appropriate studies. A similar comment applies for injection of the scleral vessels. A scoring system (the sepsis score) has been developed to aid in the identification of foals with sepsis. The sepsis score was developed with the intention of aiding identification of foals with sepsis, thereby facilitating appropriate treatment. A table for calculation of the sepsis score (the modified sepsis score) is provided in Table 19-11. Foals with a score of 12 or greater are considered to be septic, with a sensitivity of 94% in the original report. However, more recent studies, including one of 1095 foals, have found the sensitivity and specificity of the sepsis score to be less than the original report. The modified sepsis score detected sepsis with a sensitivity of 56% and a specificity of 73% using a cutoff value of 11 or more. A cutoff value of 7

yielded a sensitivity of 84% and specificity of 42%.3 These recent studies are broadly consistent with earlier studies that demonstrate that the sepsis score has limited sensitivity (67%, 95% CI 59% to 75%) and specificity (76%, 95% CI 68% to 83%) in foals less than 10 days of age. Similarly, 49% of 101 foals with positive blood cultures had a sepsis score of 11 or less, indicating a low specificity of the test. The low to moderate sensitivity of the sepsis score for detection of sepsis or bacteremia means that many foals with sepsis are incorrectly diagnosed as being nonseptic (i.e., a high false-negative rate), whereas a moderate to low specificity means that the falsepositive rate might be excessive, with a number of foals being considered septic when they are not. This is an important shortcoming of the test because accurate and prompt identification of foals with sepsis is assumed to be important for both prognostication and selection of treatment. The sepsis score might be useful in some situations, but its shortcomings should be recognized when using it to guide treatment or determine prognosis.

1859

Prematurity and Dysmaturity Detection of prematurity is important because it is a strong risk factor for development of other diseases during the immediate postpartum period. The detection of prematurity is often based on the length of gestation. However, the duration of gestation in Thoroughbred horses varies considerably, with 95% of mares foaling after a gestation of 327 to 357 days. The generally accepted “average” gestation is 349 days, with fillies having shorter gestations than do colts (348 versus 350 days) and gestation length declining by approximately 20 days, from 360 to 340 days, in Standardbred mares in New Zealand.4 Ponies have a shorter gestation (333 days, range 315 to 350 days). Therefore a diagnosis of prematurity should be based not just on gestational age but also on the results of physical, hematologic, and serum biochemical examination of the newborn. Factors helping in the determination of prematurity are listed in Table 19-4. Foals that are immature (premature) at birth typically have low birth weight and small body size, a short and silky hair coat, and laxity of the

Table 19-4  Criteria to assess stage of maturity of the newborn foal Criterion

Premature

Full term

Physical Gestational age Size Coat Fetlock

320 d Small Short and silky Overextended

Normally > 330 d Normal or large Long Normal extension

Behavior First stand First stand Suck reflex Righting reflexes

>120 min >3 h Poor Poor