Endocrinology and Systemic Diseases [1 ed.] 9783319687285, 9783319687292, 9783319687308

This work offers a comprehensive reference guide to help clinicians in the field of Internal Medicine addressing common

566 104 9MB

English Pages 495 [487] Year 2020

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Endocrinology and Systemic Diseases [1 ed.]
 9783319687285, 9783319687292, 9783319687308

Table of contents :
Series Preface
Volume Preface
Contents
About the Editors
Contributors
1 Impact of Endocrine Disorders on Typical and Atypical Cardiovascular Risk Factors
Introduction
Effects of Glucose Metabolism Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Effects of Prolactin Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Effects of Sexual Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Androgen Excess in Women and Related Metabolic Consequences
Polycystic Ovary Syndrome
Women with Monogenic Causes of Androgen Excess
Women with Monogenic Insulin Resistance
Androgen Deficiency in Men and Related Metabolic Consequences
Primary Male Hypogonadism
Secondary Male Hypogonadism
Acquired Male Hypogonadism
The Role of Androgens in Metabolic Target Tissues
Androgens, Adipose Tissue, and Lipid Metabolism
Insulin Resistance, Type 2 Diabetes Mellitus, and Androgen Status in Men and Women
Female Androgen Excess and Insulin Resistance
Male Androgen Deficiency and Insulin Resistance
Body Composition and Impact of Androgen Status in Men and Women
Female Androgen Excess on Body Composition
Male Androgen Deficiency and Body Composition
Nonalcoholic Fatty Liver Disease (NAFLD) and Male and Female Androgen Status
Female Androgen Excess and NAFLD
Male Androgen Deficiency and NAFLD
Cardiovascular Risk and Male and Female Androgen Status
Female Androgen Excess and Cardiovascular Risk
Male Androgen Deficiency and Cardiovascular Risk
Effects of Thyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Overt and Subclinical Hyperthyroidism
Overt and Subclinical Hypothyroidism
Thyroid Hormones and Cardiovascular Risk Factors
Hyperlipidemia
Insulin Resistance
Blood Pressure and Vascular Function
Thrombosis
Effects of Growth Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Growth Hormone Excess
Growth Hormone Deficiency
Effects of Adrenocorticotropic Hormone and Cortisol Disturbances on Cardiovascular Traditional and Nontraditional Cardiovascul...
Effects of Aldosterone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Aldosterone and Insulin Resistance
Aldosterone and Endothelial Function
Aldosterone and Hypertension
Aldosterone and Cardiac Structure
Aldosterone and Congestive Heart Failure
Pheochromocytoma
Effects of Parathyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors
Hyperparathyroidism
Hyperparathyroidism and Hypertension
Hyperparathyroidism Glucose Metabolism
Hyperparathyroidism and Atherosclerosis
Hypoparathyroidism
Effects of Intestinal Dysbacteriosis on Traditional and Nontraditional Cardiovascular Risk Factors
Gut Microbiota Disturbances and Hyperlipidemia
Gut Microbiota Disturbances and Obesity
Gut Microbiota Disturbances and Atherosclerosis
Gut Microbiota Disturbances and Cardiovascular Disease
Summary
Cross-References
References
2 Impact of Endocrine Disorders on Blood Pressure
Introduction
Diabetes and Metabolic Syndrome
The Crosstalk Between Diabetes and Hypertension
Pathophysiological Interactions Between Diabetes and Hypertension
The Effect of Antidiabetic Drugs in Hypertension
Thyroid
Hyperthyroidism
Hypothyroidism
Mineralocorticoids and Glucocorticoids
Mineralocorticoid and BP Physiology
Hypertensive Disorders
Primary Aldosteronism
CS
CAH
Liddle´s Syndrome (or Pseudohyperaldosteronism)
AME
Geller´s Syndrome
Mineralocorticoid Hypotension
Chronic Primary Adrenal Insufficiency (PAI)
Acute Adrenal Insufficiency
Selective Hypoaldosteronism
Pseudohypoaldosteronism (PHA)
GH and IGF-1
GH Excess (Acromegaly)
GH Deficiency (GHD)
Hyperparathyroidism and Vitamin D Deficiency
Interactions Between Ca2+/Phosphate Homeostasis and Renin-Angiotensin-Aldosterone System
Hyperparathyroidism and BP
Vitamin D and BP
Sexual Hormones
Estrogen
Testosterone
Summary
References
3 Impact of Endocrine Disorders on the Heart
Introduction
The Pituitary Gland and Cardiovascular Disease
Prolactin
Peripartum Cardiomyopathy (PPCM)
Growth Hormone
Follicle-Stimulating Hormone-Luteinizing Hormone (FSH-LH) and Testosterone
Adrenocorticotropic Hormone (ACTH)-Cortisol
Thyroid Hormone-Mediated Cardiac Disease
Thyroid Hormone-Related Coronary Artery Disease
Hyperthyroidism
Hypothyroidism
Amiodarone-Induced Thyroid Disease
Parathyroid Hormone-Related Cardiovascular Disease
Hyperparathyroidism, Vitamin D, and Calcium Homeostasis
Hypoparathyroidism
Carcinoid Heart Disease (Hart et al. 2017)
Neuroendocrine Tumors (NETs)
Carcinoid Heart Disease
Surgical Intervention
Diabetes and Heart Disease
Introduction
Classification of DM
DM and Cardiovascular Disease
Functional and Structural Alterations
Diabetic Cardiomyopathy
Cardiovascular Risk Assessment
Prevention and Management of Cardiovascular Disease
Revascularization
Arrhythmia
Heart Failure
Conclusion
Pheochromocytoma-Related Cardiovascular Disease
Spontaneous Coronary Artery Dissection
Broken Heart Syndrome
Summary
Cross-References
References
4 Impact of Endocrine Disorders on Vasculature
Introduction
Diabetes and Metabolic Syndrome
The Effect of Hyperglycemia on Vascular Physiology and Pathophysiology
The Effect of Insulin Imbalance on Vascular Physiology and Pathophysiology
Thyroid
Direct Effects of Hypothyroidism on Endothelial Dysfunction
Indirect Effects of Hypothyroidism on Endothelial Dysfunction
The Role of Hyperthyroidism on Vascular Function
Mineralocorticoids
Aldosterone Effects on Endothelial Dysfunction, Vascular Remodeling, and Stiffening
Effects of Aldosterone in Atherosclerotic Plaque Progression
Clinical Evidences
Glucocorticoids (GCs)
Clinical Evidences
Growth Hormone (GH) and Insulin-Like Growth Factor (IGF)-1
GH Excess (Acromegaly)
GH Deficiency (GHD)
Hyperparathyroidism and Vitamin D Deficiency
PTH and Vascular Homeostasis
The Vitamin D System in Vascular Biology
Fibroblast Growth Factor (FGF) 23/Klotho Axis: A New Player
Sexual Hormones
Estrogens
Testosterone
Conclusions
References
5 Impact of Endocrine Disorders on the Kidney
Introduction
Diabetes
Epidemiology
Pathogenesis
Clinical Features
Histological Findings
Therapeutic Approach
Glycemic Control
Blood Pressure Control
Obesity
Parathyroids
Thyroid
Adrenal Glands
Medulla Adrenal Disease
Pituitary Gland
Diabetes Insipidus
Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)
Sexual Hormones
Male
Female
Summary
Diabetes
Obesity
Parathyroids
Thyroid Hormones
Adrenal Glands
Medulla Adrenal Disease
Pituitary Gland
Diabetes Insipidus
Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)
Sexual Hormones
Cross-References
References
6 Impact of Endocrine Disorders on the Liver
Introduction
Liver and Thyroid Diseases
Adrenal Glands and the Liver
Adrenal Insufficiency
Adrenal Excess
Hyperaldosteronism
Adrenal Catecholamines
Pituitary Hormones and the Liver
Sex Hormones and the Liver
Estrogens
Androgens
Gonadal Dysfunction in Liver Diseases
Polycystic Ovary Syndrome (PCOS)
Summary
Cross-References
References
7 Impact of Endocrine Disorders on Gastrointestinal Diseases
Introduction
Obesity
Esophagus
Stomach
Small Intestine
Colon and Rectum
Liver
Gallbladder
Pancreas
Diabetes
Esophagus
Stomach
Lower Gastrointestinal Tract
Liver
Gallbladder
Adrenal Insufficiency
Acromegaly
Thyroid Disorders
Hyperthyroidism
Hypothyroidism
Endocrine Tumors
Pancreatic Neuroendocrine (Islet-Cell) Tumors
Carcinoid Tumors
Multiple Endocrine Neoplasia (MEN)
von Hippel-Lindau Disease
Emerging Fields
Gut Microbiota
Bile Acids as Signaling Molecules with Systemic Effects
Summary and Future Trends
References
8 Impact of Endocrine Disorders on Autoimmune Diseases
Introduction
Thyroid Disease
Rheumatological Manifestations of Thyroid Diseases
The Association of Autoimmune Thyroid Diseases with Other Rheumatic Diseases
Thyroid Disease and RA
Autoimmune Thyroid Disease and SLE
Autoimmune Thyroid Disease and Sjögren Syndrome
Diabetes
Shoulder and Hand Syndromes
Diabetes and RA
The Impact of Treatment of RA on Insulin Resistance
Diabetes and SLE
Disorders of the Hypothalamic-Hypopituitary-Adrenal (HPA) Axis
Adrenal Disorders
Acromegaly and Gigantism
Diabetes Prolactin
Hyperprolactinemia and Autoimmunity
Prolactin and SLE
Prolactin and RA
Prolactin and Sjögren Syndrome
Parathyroid Disorders
Hypoparathyroidism
Hyperparathyroidism
Parathyroid Disorders and Rheumatic Diseases
Summary
References
9 Neuroendocrine Involvement in Immune-Mediated Rheumatic Diseases
Introduction
Hormones and Immune System Interactions
Nervous System and Immune System Interactions
Clinical Conditions in Which Alterations of Neuroendocrine System Was Recognized
Rheumatoid Arthritis
Polymyalgia Rheumatica/Giant Cell Arteritis
Systemic Lupus Erythematosus
Sjögren Syndrome
A Final Note on the Adipose Tissue and Adipokines
Conclusion
References
10 Neurological Manifestations of Endocrine Disorders
Introduction
The Endocrine System
Neurological Manifestations of Different Endocrine Disorders
Pituitary Disorders
Anatomy and Physiology
Types of Pituitary Lesions
Neurological Manifestations of Pituitary Lesions
Neurological Symptoms Associated with Dysfunction of Direct-Acting Hormones of the Anterior Pituitary Gland
Growth Hormone (GH)
Prolactin (PRL)
Vasopressin (Antidiuretic Hormone)
Diagnosis and Treatment
Thyroid Disorders
Anatomy and Physiology
Types of Lesion
Neurological Manifestations of Thyroid Disorders
Hypothyroidism
Encephalopathy
Cognitive Impairment
Peripheral Neuropathy
Myopathy
Other Manifestations of Hypothyroidism
Hyperthyroidism
Cognitive Impairment and Psychiatric Symptoms
Peripheral Neuropathy
Muscle Involvement
Tremor and Other Abnormal Movements
Ophthalmopathy
Other Manifestations of Hyperthyroidism
Hashimoto Encephalopathy
Adrenal Disorders
Anatomy and Physiology
Types of Lesion
Neurological Manifestations of Adrenal Disorders
Cushing´s Syndrome
Pheochromocytoma
Adrenal Insufficiency
Adrenoleukodystrophy (Adrenoleukodystrophy-Adrenomyeloneuropathy Complex)
Mineral Metabolism and Parathyroid Disorders
Anatomy and Physiology
Types of Lesion
Neurological Manifestations of Parathyroid Disorders
Hyperparathyroidism
Fahr Syndrome
Hypocalcemia
Diagnosis and Treatment
Gonadal Disorders
Anatomy and Physiology
Types of Lesion
Neurological Manifestations of Gonadal Disorders
Estrogen Deficiency During Menopause
Testosterone Deficiency in Elderly Patients
Paraneoplastic Neurological Symptoms Associated with Gonadal Tumors
Fragile X Syndrome
Kennedy Disease
Pituitary and Hypothalamic Disorders
Obesity
Neurological Manifestations of Obesity
Migraine
Cognitive Impairment
Polyneuropathy
Diabetes Mellitus
Neurological Manifestations of Diabetes Mellitus
Stroke
Cognitive Impairment
Diabetic Neuropathy
Symmetric Distal Sensory or Sensorimotor Polyneuropathy
Small-Fiber Polyneuropathy
Autonomic Neuropathy
Focal Neuropathies
Diabetic Amyotrophy
Summary
Cross-References
References
11 Endocrine Disorders and Psychiatric Manifestations
Introduction
General Mechanisms That Link Psychiatric Manifestations and Endocrine Disorders
General Psychiatric Manifestations of Endocrine Diseases
Pituitary Disorders
Hypopituitarism
Cushing´s Disease
Acromegaly
Hyperprolactinemia
Gonadal Disorders
Male Hypogonadism
Female Hypogonadism
Precocious Puberty
Adrenal Disorders
Addison´s Disease
Primary Aldosteronism
Pheochromocytoma and Paraganglioma
Congenital Adrenal Hyperplasia
Thyroid Disorders
Hypothyroidism
Subclinical Hypothyroidism
Hyperthyroidism
Subclinical Hyperthyroidism
Autoimmune Thyroiditis and Hashimoto´s Encephalopathy
Thyroid Cancer
Parathyroid Disorders
Primary Hyperparathyroidism
Primary Hypoparathyroidism
Polycystic Ovary Syndrome
Diabetes Mellitus
Obesity
Effects of Bariatric Surgery
Hormonal Treatment and Psychiatric Disease
Psychiatric Diseases Inducing Endocrine Pathology
Summary
Cross-References
References
12 Impact of Endocrine Disorders in Critically Ill Patients
Introduction
Dysglycemia in Critically Illness
General Concepts and Assessment
Pathogenetic Mechanisms and Consequences
Dysglycemia in Different Critically Ill Patients
Low-T3 Syndrome or Non-thyroidal Illness Syndrome
General Concepts and Assessment
Primary Hypothyroidism in Critically Illness
LowT3S Management and Therapy
References
13 Impact of Endocrine Disorders in the Elderly
Introduction
Hypofiso-Gonadal Function
Women
Symptoms of Menopause
Hot Flashes or Vasomotor Symptoms
Vaginal Dryness
Depression
Sleep Disturbance
Neurocognitive Changes
Osteoporosis
Body Composition
Cardiovascular Disease
Estrogen Replacement Therapy
Men
Change in Gonadotropins
Serum Testosterone, Sex Hormone-Binding Globulin
Clinical Consequences of Testosterone Decline
Bone Mineral Density and Fractures
Cardiovascular Disease and Body Composition
Neuropsychiatric Functions
Decline in Spermatogenesis
Testosterone Replacement Therapy
Other Hypothalamic-Neuro/Adeno-hypofisary Axis
GH/IGF-1
Thyroid-Stimulating Hormone
Vasopressin/Antidiuretic Hormone
Thyroid Function
Adrenal Cortex
Cortisol
Dehydroepiandrosterone
Aldosterone
Adipose Tissue and Skeletal Muscle
Sarcopenia
Adipose Tissue Depot
Overnutrition
Undernutrition
Management of Sarcopenia and Dysfunctional Changes in Body Adiposity
Physical Activity
Healthy Diet
Overnutrition
Undernutrition
Hormone-Dependent Calcium Balance
Calcium Balance
Vitamin D
Parathyroid Hormone
Impact of Hormonal Disorders in Persons Aged Above 75 Years
Summary
Cross-References
References
14 Impact of Endocrine Disorders on Skin Disorders
Introduction
Pituitary Gland
Hypopituitarism
Hyperpituitarism
Acromegaly
McCune-Albright Syndrome
Hyperprolactinemia
Prolactinoma
Thyroid Gland
Congenital Hypothyroidism
Hypothyroidism
Hyperthyroidism
Graves´ Disease
Parathyroid Gland
Hypoparathyroidism
Hyperparathyroidism
Adrenal Glands
Hyperadrenalism
Cushing´s Syndrome
Pseudo-Cushing´s Disease
Addison´s Disease
Pheochromocytoma
Hypogonadism
Excessive Androgen Production
Polycystic Ovarian Syndrome (PCOS)
Pancreas
Diabetes Mellitus (DM)
Necrobiosis Lipoidica Diabeticorum
Granuloma Annulare
Diabetic Bullae
Scleredema Diabeticorum
Cutaneous Infections
The Diabetic Foot
Acanthosis Nigricans
Diabetic Dermopathy
Acquired Perforating Dermatoses
Eruptive Xanthomatosis
Vitiligo
Acrochordons (Skin Tags)
Yellow Skin and Nails
Generalized Pruritus
Ichthyosiform Skin
Lichen Planus
Rubeosis Faciei
Palmar Erythema
Nail Bed Erythema
Pigmented Purpura
Cutaneous Reactions to Diabetic Treatment
Adverse Skin Reactions to Insulin
Oral Hypoglycemic Agents
Polyendocrine Disorders
Multiple Endocrine Neoplasia (MEN) Syndromes
Autoimmune Polyglandular Syndromes
Genetic Syndromes
Carney Complex
21-Hydroxylase Deficiency
Neuroendocrine Tumors
Glucagonoma Syndrome
Carcinoid Syndrome
Merkel Cell Carcinoma
Summary
References
15 The Role of Environmental Pollution in Endocrine Diseases
Introduction
Insulin Resistance, Obesity, Type 2 Diabetes
Type 1 Diabetes
Thyroid Diseases
Reproductive Abnormalities
Endocrine Disruptors and Cancer
Endocrine Effects of Radiofrequency Electromagnetic Fields
Summary and Future Trends
References
Index

Citation preview

Endocrinology Series Editor: Andrea Lenzi Series Co-Editor: Emmanuele A. Jannini

Piero Portincasa Gema Frühbeck Hendrik M. Nathoe  Editors

Endocrinology and Systemic Diseases

Endocrinology Series Editor Andrea Lenzi Department of Experimental Medicine Section of Medical Pathophysiology Food Science and Endocrinology Sapienza University of Rome Rome, Italy Series Co-Editor Emmanuele A. Jannini Department of Systems Medicine University of Rome Tor Vergata Rome, Italy

Within the health sciences, Endocrinology has an unique and pivotal role. This old, but continuously new science is the study of the various hormones and their actions and disorders in the body. The matter of Endocrinology are the glands, i.e. the organs that produce hormones, active on the metabolism, reproduction, food absorption and utilization, growth and development, behavior control, and several other complex functions of the organisms. Since hormones interact, affect, regulate and control virtually all body functions, Endocrinology not only is a very complex science, multidisciplinary in nature, but is one with the highest scientific turnover. Knowledge in the Endocrinological sciences is continuously changing and growing. In fact, the field of Endocrinology and Metabolism is one where the highest number of scientific publications continuously flourishes. The number of scientific journals dealing with hormones and the regulation of body chemistry is dramatically high. Furthermore, Endocrinology is directly related to genetics, neurology, immunology, rheumatology, gastroenterology, nephrology, orthopedics, cardiology, oncology, gland surgery, sexology and sexual medicine, psychology, psychiatry, internal medicine, and basic sciences. All these fields are interested in updates in Endocrinology. The aim of the MRW in Endocrinology is to update the Endocrinological matter using the knowledge of the best experts in each section of Endocrinology: basic endocrinology, neuroendocrinology, endocrinological oncology, pancreas with diabetes and other metabolic disorders, thyroid, parathyroid and bone metabolism, adrenals and endocrine hypertension, sexuality, reproduction, and behavior. More information about this series at http://www.springer.com/series/14021

Piero Portincasa • Gema Fru¨hbeck • Hendrik M. Nathoe Editors

Endocrinology and Systemic Diseases With 48 Figures and 20 Tables

Editors Piero Portincasa Department of Biomedical Sciences and Human Oncology University of Bari “Aldo Moro”, Clinica Medica “Augusto Murri” Bari, Italy

Gema Frühbeck Department of Endocrinology & Nutrition University of Navarra, CIBEROBN, IdiSNA, Clinica Universidad de Navarra Pamplona, Spain

Hendrik M. Nathoe Department of Cardiology University Medical Center Utrecht, The Netherlands

ISSN 2510-1927 ISSN 2510-1935 (electronic) ISBN 978-3-319-68728-5 ISBN 978-3-319-68729-2 (eBook) ISBN 978-3-319-68730-8 (print and electronic bundle) https://doi.org/10.1007/978-3-319-68729-2 © Springer Nature Switzerland AG 2021 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. This Springer imprint is published by the registered company Springer Nature Switzerland AG. The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland

Series Preface

Is there an unmet need for a new MRW series in Endocrinology and Metabolism? It might not seem so! The vast number of existing textbooks, monographs and scientific journals suggest that the field of hormones (from genetic, molecular, biochemical and translational to physiological, behavioral, and clinical aspects) is one of the largest in biomedicine, producing a simply huge scientific output. However, we are sure that this new series will be of interest for scientists, academics, students, physicians and specialists alike. The knowledge in Endocrinology and Metabolism limited to the two main (from an epidemiological perspective) diseases, namely hypo/hyperthyroidism and diabetes mellitus, now seems outdated and perhaps closer to the practical interests of the general practitioner than to those of the specialist. This has led to endocrinology and metabolism being increasingly considered as a subsection of internal medicine rather than an autonomous specialization. But endocrinology is much more than this. We are proposing this series as the manifesto for Endocrinology 2.0, embracing the fields of medicine in which hormones play a major part but which, for various historical and cultural reasons, have thus far been “ignored” by endocrinologists. Hence, this MRW comprises “traditional” (but no less important or investigated) topics: from the molecular actions of hormones to the pathophysiology and management of pituitary, thyroid, adrenal, pancreatic and gonadal diseases, as well as less usual and common arguments. Endocrinology 2.0 is, in fact, the science of hormones, but it is also the medicine of sexuality and reproduction, the medicine of gender differences and the medicine of well-being. These aspects of Endocrinology have to date been considered of little interest, as they are young and relatively unexplored sciences. But this is no longer the case. The large scientific production in these fields coupled with the impressive social interest of patients in these topics is stimulating a new and fascinating challenge for Endocrinology. The aim of the MRW in Endocrinology is thus to update the subject with the knowledge of the best experts in each field: basic endocrinology, neuroendocrinology, endocrinological oncology, pancreatic disorders, diabetes and other metabolic disorders, thyroid, parathyroid and bone metabolism, adrenal and endocrine

v

vi

Series Preface

hypertension, sexuality, reproduction and behavior. We are sure that this ambitious aim, covering for the first time the whole spectrum of Endocrinology 2.0, will be fulfilled in this vast Springer MRW in Endocrinology Series. Andrea Lenzi Emmanuele A. Jannini

Volume Preface

Endocrinology and Systemic Diseases combines common aspects of clinical endocrinology with major topics of internal medicine and provides a comprehensive outlook of the complex and bi-directional relationship between hormonal homeostasis and a variety of organ dysfunctions. The editors are fully aware of the exponential expansion of knowledge in biomedicine, which requires continuous multidisciplinary approaches across specialists, disciplines, health policies, and established evidence-based guidelines. On the other hand, the risk deriving from overspecialization could lead to the lack of an integrative approach, increasing diagnosis mistakes and health costs. The daily need of individual practitioners is to address multiple, complex, and often overlapping diagnoses, available diagnostic and therapeutic capacities, confounding subclinical manifestations, cross-links between dysfunctions in different systems, and integration of single cases with ongoing guidelines in a rapidly evolving scientific knowledge. In this first edition, experts at the forefront of knowledge in their specific fields focus on the multifactorial mechanisms linking endocrinological disorders and systemic abnormalities, which include liver and gastrointestinal diseases; heart and cardiovascular diseases; kidney, autoimmune, neuroendocrine and rheumatic disorders; neurological and psychiatric manifestations; critically ill conditions; elderly patients; and dermatological diseases. Because of close attention to the role of environment on human health, a chapter also deals with the role of environmental pollution in endocrine diseases. This first edition of Endocrinology and Systemic Diseases therefore intercepts the rapid change of knowledge in endocrine aspects of clinical medicine and provides a useful tool to adequately manage the altered hormonal balance frequently linked with several clinical conditions. We trust that the book will be helpful and informative, and therefore will support and improve the continuous medical education of trainees and MD specialists, for a full management of complex clinical cases.

vii

viii

Volume Preface

We thank all authors who contributed with both enthusiasm and dedication to the impact of the book. Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology University of Bari Medical School Bari, Italy Senior Consultant of Department of Endocrinology and Nutrition University Clinic of Navarra CIBEROBN, Avda. Pio XII, 36 Pamplona, Spain University Medical Center Utrecht Utrecht, The Netherlands January 2021

Prof. Piero Portincasa

Prof Gema Frühbeck

Prof. Hendrik M. Nathoe Editors

Contents

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . M. Perticone and F. Perticone

1

2

Impact of Endocrine Disorders on Blood Pressure . . . . . . . . . . . . Federico Carbone, Aldo Bonaventura, Alessandra Vecchiè, Matteo Casula, Luca Liberale, Franco Dallegri, and Fabrizio Montecucco

29

3

Impact of Endocrine Disorders on the Heart . . . . . . . . . . . . . . . . . Marcel Th. B. Twickler, Jakub Regieli, Remco Grobben, Einar A. Hart, Margot E. Tesselaar, Ronald C. A. Meijer, Steven A. J. Chamuleau, and Hendrik M. Nathoe

59

4

Impact of Endocrine Disorders on Vasculature . . . . . . . . . . . . . . . Federico Carbone, Aldo Bonaventura, Luca Liberale, Alessandra Vecchié, Matteo Casula, Franco Dallegri, and Fabrizio Montecucco

97

5

Impact of Endocrine Disorders on the Kidney . . . . . . . . . . . . . . . . Silvia Matino, Francesco Pesce, Michele Rossini, Giuseppina D’Ettorre, Alessandro Mascolo, and Loreto Gesualdo

123

6

Impact of Endocrine Disorders on the Liver . . . . . . . . . . . . . . . . . Agostino Di Ciaula, Grigorios Christidis, Marcin Krawczyk, Frank Lammert, and Piero Portincasa

157

7

Impact of Endocrine Disorders on Gastrointestinal Diseases . . . . . Agostino Di Ciaula, David Q. H. Wang, Thomas Sommers, Antony Lembo, and Piero Portincasa

179

8

Impact of Endocrine Disorders on Autoimmune Diseases . . . . . . . Hagit Peleg and Eldad Ben-Chetrit

227

9

Neuroendocrine Involvement in Immune-Mediated Rheumatic Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Maurizio Cutolo and Amelia Chiara Trombetta

263

ix

x

Contents

10

Neurological Manifestations of Endocrine Disorders . . . . . . . . . . . Pablo Irimia Sieira, Ane Mínguez Olaondo, Eduardo Martínez-Vila, and Martin Ruttledge

281

11

Endocrine Disorders and Psychiatric Manifestations . . . . . . . . . . . Javier Salvador, Gala Gutierrez, María Llavero, Javier Gargallo, Javier Escalada, and Judith López

311

12

Impact of Endocrine Disorders in Critically Ill Patients Gabriella Garruti, Ana F. Pina, M. Paula Machedo, and Francesco Giorgino

........

347

13

Impact of Endocrine Disorders in the Elderly . . . . . . . . . . . . . . . . P. Sunsundegui, V. Santesteban-Moriones, and M. F. Landecho

365

14

Impact of Endocrine Disorders on Skin Disorders . . . . . . . . . . . . . Domenico Bonamonte and Angela Filoni

399

15

The Role of Environmental Pollution in Endocrine Diseases . . . . . Agostino Di Ciaula and Piero Portincasa

435

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

467

About the Editors

Prof. Piero Portincasa (M.D., Ph.D. [Utrecht], dr. H. Causa) is Full Professor of Internal Medicine and Chief of the Academic Division of Internal Medicine “A. Murri,” Department of Biomedical Sciences and Human Oncology at the University “Aldo Moro” Medical School and of the Unit of Internal Medicine “A. Pende,” Regional Hospital Policlinico in Bari, Italy. He is director of the 4-year residency program in geriatrics. The activities as internist, teacher, and scientist combine both basic and clinical research in the field of dyslipidaemia and metabolic disorders, liver steatosis and gallstone disease, and rare autoinflammatory diseases (Familial Mediterranean Fever). His teaching and research group deals with aspects of food digestion and intolerances, gastrointestinal motility, and application of standard and novel diagnostic tests in gastrointestinal and liver diseases. He is author of more than 500 international papers and several books and chapters. Prof. Portincasa worked as University Research Fellow at Guy’s Hospital, London, UK (1985–87); as Clinician and Ph.D. student at the Academic Hospital in Utrecht, the Netherlands (1993–95); and as Clinical Fellow of the National Research Centre (CNR) at Harvard Medical School, Beth Israel Deaconess Medical Centre, Boston, USA (2005). He served as Councillor of the European Society for Clinical Investigation, ESCI (2008–11), President of ESCI (2011–15), President of Foundation of European Journal of Clinical Investigation (2015–18), and since 2018 is President of ESCI Trust. He was the organizer of the 2010 Annual scientific Meeting of ESCI in Bari and he will organize again the next ESCI meeting in 2021, in Bari. Prof. Portincasa

xi

xii

About the Editors

holds the Laurea H. Causa at the Universities “C. Davila,” Bucharest (2013), “Iuliu Hatieganu” ClujNapoca (2013), and “George Emil Palade” TarguMures (2018), Romania. He is member of the Apulian Academy of Sciences, past Councillor and Secretary (2013–17) of the Italian Society Internal Medicine, and recipient of the prestigious ESCI Albert Struyvenberg Medal in 2018. He also holds the Fellowship of the American Gastroenterological Association (AGAF). Prof. Portincasa is active in international relations as Erasmus+ coordinator in Bari and worldwide as member of several international scientific societies and journal editorial boards. He is mentoring undergraduate and postgraduate students of medicine, biology, and pharmacy also involved in the Horizon 2020 Foie Gras project. Prof. Gema Frühbeck (RNutr, M.D., Ph.D.) combines everyday work in the clinical setting as Director of the Obesity Area and Senior Consultant in the Department of Endocrinology and Nutrition at the Clínica Universidad de Navarra (Pamplona, Spain) with basic research as Head of the Metabolic Research Laboratory at the University of Navarra and Project Leader of CIBEROBN (Centre of Excellence Network in Obesity and Nutrition of the Spanish Institute of Health). Her group has taken the step forward of routinely using body composition measurements in the clinical assessment of patients in order to analyze the relevance of body fat – beyond body mass index – in comorbidity development. In 1997, she worked as Postdoctoral Fellow of the Royal Society in Prof. Andrew Prentice’s Energy Group at the MRC Dunn Clinical Nutrition Centre in Cambridge, UK. In 2009, she was unanimously voted PresidentElect of the European Association for the Study of Obesity. Prof. Frühbeck is also a recipient of the Nutrition Society’s Silver Medal, UK (2000), “Sir David Cuthbertson Medal” (2005), Lilly Distinguished Career Award in Endocrinology and Nutrition (2011), and Albert Struyvenberg Medal (2013). From 2012 to 2015 Prof. Frühbeck served as President of the European Association for the Study of Obesity. She has also served as Councillor of the European Society for Clinical Investigation (2014–2018).

About the Editors

xiii

Hendrik M. Nathoe was born in Surinam, South America, and moved to the Netherlands in 1983. He studied medicine at the Erasmus Medical Center Rotterdam and graduated summa cum laude in 1991. Prof. Nathoe received his Ph.D. degree in 2004 from the University of Utrecht under supervision of Prof. Diederick Grobbee and Prof. Peter de Jaegere His thesis was entitled Coronary revascularization: Stentimplantation, on-pump or off-pump bypass surgery. After his training he was appointed in 2006 at the University Medical Center Utrecht as General Cardiologist and Associate Professor of Cardiology. Since 2018, he has been Medical Head of the Center for Cardiovascular Disease, the care facility of the UMC Utrecht research focus “Circulatory Health”. Here, he collaborates with other vascular specialists to optimize the care of multidisciplinary vascular patients. In 2019, this center was accredited as one of the first in Europe by the European Society of Cardiology for primary and secondary prevention and sports medicine. Since 2020, he has been appointed as Medical Head of the outpatient clinic in the Department of Cardiology at UMC Utrecht. Prof. Nathoe was involved in the governance of the European Society for Clinical Investigation (ESCI) since 2013 as Secretary Treasurer and President. He organized the annual scientific meeting of ESCI in 2014 in Utrecht. He (co)-authored 125 peer-reviewed articles of whom one as first author in the New England Journal of Medicine. He supervised 6 Ph.D. students. He is member of many steering committees such as SMART study, CHASE investigators, Octopus study, BEACON trial, DECS study, and MARC study. Prof. Nathoe was involved in the 2019 European Society of Cardiology guidelines on “the management of chronic coronary syndromes.” Since 2020, he is Editor-in-Chief of the European Journal of Clinical Investigation.

Contributors

Eldad Ben-Chetrit Rheumatology Unit, Department of Medicine, HadassahHebrew University Medical Center, Jerusalem, Israel Domenico Bonamonte Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy Aldo Bonaventura First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy Federico Carbone First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy Matteo Casula First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy Steven A. J. Chamuleau University Medical Center Utrecht, Utrecht, The Netherlands Grigorios Christidis Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany Maurizio Cutolo Research Laboratory and Clinical Division of Rheumatology, Department of Internal Medicine, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy Giuseppina D’Ettorre Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Franco Dallegri First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy IRCCS Ospedale Policlinico San Martino – Italian Cardiovascular Network, Genoa, Italy

xv

xvi

Contributors

Agostino Di Ciaula Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy Division of Internal Medicine, Hospital of Bisceglie (BAT), ASL BAT, Bisceglie, Italy International Society of Doctors for Environment – ISDE, Basel, Switzerland Javier Escalada Department of Endocrinology and Nutrition, University Clinic of Navarra, Pamplona, Spain CIBERObn, Institute Carlos III, Madrid, Spain Angela Filoni Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy Javier Gargallo Department of Endocrinology and Nutrition, University Clinic of Navarra, Pamplona, Spain Gabriella Garruti Section of Internal Medicine, Endocrinology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Medical School, Bari, Italy Loreto Gesualdo Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Francesco Giorgino Section of Internal Medicine, Endocrinology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Medical School, Bari, Italy Remco Grobben University Medical Center Utrecht, Utrecht, The Netherlands Gala Gutierrez Department of Endocrinology and Nutrition, University Clinic of Navarra, Pamplona, Spain Einar A. Hart University Medical Center Utrecht, Utrecht, The Netherlands Pablo Irimia Sieira Departamento de Neurología, Clínica Universidad de Navarra, Pamplona, Spain Marcin Krawczyk Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany Laboratory of Metabolic Liver Diseases, Center for Preclinical Research, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland Frank Lammert Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany M. F. Landecho General Health Check-up Unit, Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain

Contributors

xvii

Antony Lembo Division of Gastroenterology, Beth Israel and Deaconess Center, Harvard Medical School, Boston, MA, USA Luca Liberale First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland María Llavero Department of Endocrinology and Nutrition, University Clinic of Navarra, Pamplona, Spain Judith López Department of Endocrinology and Nutrition, University Hospital of Canarias and University of La Laguna, Tenerife, Spain Ane Mínguez Olaondo Departamento de Neurología, Clínica Universidad de Navarra, Pamplona, Spain M. Paula Machedo Chronic Diseases Research Center, NOVA Medical School/ Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal Eduardo Martínez-Vila Departamento de Neurología, Clínica Universidad de Navarra, Pamplona, Spain Alessandro Mascolo Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Silvia Matino Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Ronald C. A. Meijer Department of Cardiothoracic Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands Fabrizio Montecucco Ospedale Policlinico San Martino, Genoa, Italy First Clinic of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), Department of Internal Medicine, University of Genoa, Genoa, Italy IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Nework, Genoa, Italy Hendrik M. Nathoe Department of Cardiology, University Medical Center, Utrecht, The Netherlands Hagit Peleg Rheumatology Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel M. Perticone Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy F. Perticone Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy

xviii

Contributors

Francesco Pesce Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Ana F. Pina Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal Piero Portincasa Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Clinica Medica “Augusto Murri”, Bari, Italy Jakub Regieli Private cardiology clinic, Amsterdam, The Netherlands Michele Rossini Department of Emergency and Organ Transplantation (DETO), Nephrology, Dialysis and Kidney Transplantation Unit, University of Bari “Aldo Moro”, Bari, Italy Martin Ruttledge Neurology Department, Beaumont Hospital, Dublin 9, Ireland Javier Salvador Department of Endocrinology and Nutrition, University Clinic of Navarra, Pamplona, Spain CIBERObn, Institute Carlos III, Madrid, Spain V. Santesteban-Moriones General Health Check-up Unit, Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain Department of Nutrition, Food Sciences and Physiology, University of Navarra, Pamplona, Spain Centre for nutrition research, University of Navarra, Pamplona, Spain CIBER fisiopatologia de la obesidad y nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain Thomas Sommers Division of Gastroenterology, Beth Israel and Deaconess Center, Harvard Medical School, Boston, MA, USA P. Sunsundegui General Health Check-up Unit, Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain Margot E. Tesselaar Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands Amelia Chiara Trombetta Research Laboratory and Clinical Division of Rheumatology, Department of Internal Medicine, University of Genova, IRCCS San Martino Polyclinic Hospital, Genova, Italy Marcel Th. B. Twickler Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Edegem, Belgium AZ Monica, Antwerp/Deurne, Belgium

Contributors

xix

Alessandra Vecchiè First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy David Q. H. Wang Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular Risk Factors M. Perticone and F. Perticone

Contents Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Glucose Metabolism Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Prolactin Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Sexual Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Androgen Excess in Women and Related Metabolic Consequences . . . . . . . . . . . . . . . . . . . . . . . . Androgen Deficiency in Men and Related Metabolic Consequences . . . . . . . . . . . . . . . . . . . . . . . The Role of Androgens in Metabolic Target Tissues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Insulin Resistance, Type 2 Diabetes Mellitus, and Androgen Status in Men and Women . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Body Composition and Impact of Androgen Status in Men and Women . . . . . . . . . . . . . . . . . . . Nonalcoholic Fatty Liver Disease (NAFLD) and Male and Female Androgen Status . . . . . . Cardiovascular Risk and Male and Female Androgen Status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Thyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Overt and Subclinical Hyperthyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Overt and Subclinical Hypothyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Thyroid Hormones and Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

3 4 4 5 5 7 8 9 10 10 11 11 13 14 14

M. Perticone (*) Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy e-mail: [email protected] F. Perticone Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy e-mail: [email protected] © Springer Nature Switzerland AG 2021 P. Portincasa et al. (eds.), Endocrinology and Systemic Diseases, Endocrinology, https://doi.org/10.1007/978-3-319-68729-2_1

1

2

M. Perticone and F. Perticone

Effects of Growth Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Growth Hormone Excess . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Growth Hormone Deficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Adrenocorticotropic Hormone and Cortisol Disturbances on Cardiovascular Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Aldosterone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aldosterone and Insulin Resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aldosterone and Endothelial Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aldosterone and Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aldosterone and Cardiac Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aldosterone and Congestive Heart Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Parathyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hyperparathyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hypoparathyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Effects of Intestinal Dysbacteriosis on Traditional and Nontraditional Cardiovascular Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Gut Microbiota Disturbances and Hyperlipidemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Gut Microbiota Disturbances and Obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Gut Microbiota Disturbances and Atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Gut Microbiota Disturbances and Cardiovascular Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cross-References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

16 16 18 19 20 20 21 21 22 22 23 23 24 25 25 25 26 26 26 27 27

Abstract

Specific factors, both genetic and environmental, modifiable and nonmodifiable, increase the risk of developing cardiovascular (CV) diseases. Traditional cardiovascular risk factors (e.g., arterial hypertension, dyslipidemia, smoking, obesity, etc.) are strongly associated with both morbidity and mortality and can be improved through lifestyle modification and/or pharmacologic treatment. The so-called “nontraditional” cardiovascular risk factors include chronic inflammation and its markers, such as C-reactive protein, homocysteine, oxidative stress or endothelial dysfunction, lipoprotein (Lp) a, psychosocial factors (i.e., environmental stress and responsiveness to stress), plasma insulin levels and markers of insulin resistance (IR), uric acid, chronic kidney disease (CKD), and activation of the renin-angiotensin-aldosterone system (RAAS). Normal endocrine function is essential for CV health. Disorders of the endocrine system, both hyper- and hypofunction, exert multiple effects on the CV system. Keywords

Cardiovascular risk factors · Endocrine disorders · Hypertension · Insulin resistance · Dyslipidemia · Endothelial dysfunction

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

3

Introduction Population-based studies, such as the Framingham Heart Study, have established that specific factors, both genetic and environmental, modifiable and nonmodifiable, increase the risk of developing cardiovascular (CV) diseases (Dawber et al. 1951). The 27th Bethesda Conference: Matching the Intensity of Risk Factor Management with the Hazard for Coronary Disease Events has classified proposed CV risk factors into four categories based on descending levels of evidence to support the efficacy of direct management in reducing CV morbidity and mortality. Most attention in the literature has focused on the traditional risk factors in categories I and II (Table 1). These risk factors (e.g., arterial hypertension, dyslipidemia, smoking, obesity, etc.) are strongly associated with CV morbidity and mortality and can be improved through lifestyle modification and/or pharmacologic treatment. The so-called “nontraditional” CV risk factors, such as those in category III (Table 1), have been identified based on more recent studies of the pathogenesis of atherosclerosis and atherothrombotic CV events. These include chronic inflammation and its markers, such as C-reactive protein, homocysteine, oxidative stress or endothelial dysfunction, lipoprotein (Lp) a, psychosocial factors (i.e., environmental stress and responsiveness to stress), plasma insulin levels and markers of insulin resistance (IR), uric acid, chronic kidney disease (CKD), and activation of the reninangiotensin-aldosterone system (RAAS), which is in part a function of polymorphisms in genes for components of the system, such as angiotensinogen and the angiotensin II, type 1 (AT1) receptor. Endocrine disorders, involving many organs and systems, have several effects on both traditional and nontraditional CV risk factors. The following paragraphs will analyze all the endocrine disorders impacting on CV risk. Normal endocrine function is essential for CV health. Disorders of the endocrine system, both hyper- and hypofunction, exert multiple effects on the CV system.

Table 1 Proposed risk factors categories I. Risk factors for which interventions have been proved to reduce the incidence of coronary artery disease events II. Risk factors for which interventions are likely, based on our current pathophysiologic understanding and on epidemiologic and clinical trial evidence, to reduce the incidence of coronary artery disease events III. Risk factors clearly associated with an increase in coronary artery disease risk, which, if modified, might lower the incidence of coronary artery disease events IV. Risk factors associated with an increased risk but that cannot be modified or whose modification would be unlikely to change the incidence of coronary disease events Reprinted from the 27th Bethesda Conference: Matching the Intensity of Risk Factor Management with the Hazard for Coronary Disease Events. 1995 Sep 14–15. J Am Coll Cardiol 1996; 27:957–1047. Copyright © 1996 Elsevier Science

4

M. Perticone and F. Perticone

Effects of Glucose Metabolism Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Diabetes mellitus (DM) is the most common manifestation of glucose metabolism alterations, and it is both a CV risk factor and a CV disease per se. It is associated with long-term micro- and macrovascular complications. Coronary and peripheral vascular atherosclerotic disease is present in more than 50% of patients with DM, occurring at an earlier age and in a more diffuse pattern than in nondiabetic patients. Vascular disease is the primary cause of death in 80% of individuals with DM. People with DM with no prior history of heart disease have the same risk for myocardial infarction as nondiabetic patients with a known prior history of heart disease. In addition to accelerated atherosclerosis, these patients often demonstrate endothelial dysfunction secondary to early development of coronary artery disease, impaired nitric oxide release, increased serum levels of free fatty acids, and advanced end products of glycosylation. DM leads to impaired regeneration after vascular injury and impaired arteriogenesis. Dyslipidemia in patients with DM is predominantly characterized by increased triglycerides and low high-density lipoprotein level; classically, the low-density lipoprotein (LDL) level is not elevated, but the LDL particles tend to be smaller, denser, and more susceptible to oxidation, leading to increased atherogenicity. Hypercoagulability secondary to increased levels of fibrinogen and plasminogen activator inhibitor type I and enhanced platelet aggregation is present. Diabetic cardiomyopathy, which is a distinct entity from ischemic cardiomyopathy, is often seen in patients with DM and may be related to impaired energy utilization and diastolic dysfunction. Autonomic nervous system dysfunction may produce increased sympathetic tone at baseline and may contribute to silent myocardial ischemia, impaired heart rate variability, and poor prognosis (Haffner et al. 1998).

Effects of Prolactin Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Prolactin (PRL) is synthesized and secreted by lactotroph cells of the anterior pituitary gland and stimulates lactation in the postpartum period. PRL is tonically inhibited by hypothalamic dopamine. PRL levels are physiologically elevated in pregnancy, the postpartum period, and in states of stress. Pathologic hyperprolactinemia may be caused by decreased dopaminergic inhibition or by PRL secretion from benign pituitary adenomas (prolactinomas). The prevalence of hyperprolactinemia ranges from 0.4% in the general adult population to 9% in women with reproductive disorders. Although hyperprolactinemia itself does not have clear effects on the CV system, there is a possible association between long-term treatment with dopamine agonists and cardiac valve abnormalities. Dopamine agonists, including cabergoline, bromocriptine, and quinagolide, are the primary treatment for prolactinomas. High doses and long duration of therapy

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

5

with dopamine agonists have been associated (in Parkinson’s disease) with an increased risk of regurgitant valve disease. Although doses used for prolactinoma therapy are much lower than those used for Parkinson’s disease, patients with prolactinoma may be treated for decades. This treatment duration raises concern for increased risk of valvulopathy, including tricuspid regurgitation, mitral regurgitation, and aortic regurgitation (Valassi et al. 2010). Although most reports do not show an association between the use of dopamine agonists and cardiac valve disease, clinicians are advised to use the lowest possible doses of dopamine agonists. Echocardiographic monitoring should be considered, especially in patients requiring long-term and/or higher-dose therapy, and those with underlying heart or valvular disease.

Effects of Sexual Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Androgens play a major role in human metabolic health and disease. Female androgen excess (AE) and male androgen deficiency (AD) exhibit overlapping metabolic phenotypes, highlighting the complexity of the role of androgen in metabolism. Effects of androgens on adipose tissue and muscle may largely be governed by circulating serum and tissue-specific concentrations, with a narrow physiological window in both sexes, outside of which disturbances in metabolism and body composition are observed. In healthy women, low androgen concentrations and elevated estrogens lead to predominant gynecoid fat distribution and reduced metabolic risk; at circulating androgen levels observed in severe female AE and male AD, preferential accumulation of central and visceral adiposity is observed, while at higher androgen concentrations seen in healthy men, this effect is dissipated by increasing lean body mass, muscle bulk, and reducing fat mass.

Androgen Excess in Women and Related Metabolic Consequences Disturbances in androgen metabolism secondary to gonadal, adrenal, or hypothalamic-pituitary disease lead to alterations of circulating androgen concentrations and result in reproductive and metabolic complications. In women, polycystic ovary syndrome (PCOS), a triad of ovulatory dysfunction, polycystic ovarian morphology, and AE, represents the most common endocrine disorder. In men, disturbances of gonadal function most commonly result in hypogonadism and consequent AD, which can be inherited or acquired by disease, obesity, medications, or the aging process. Interestingly, female AE and male AD are associated with a similar adverse metabolic phenotype, including obesity, IR, increased prevalence of type 2 DM, nonalcoholic fatty liver disease (NAFLD), and CV disease (Zarotsky et al. 2014).

6

M. Perticone and F. Perticone

Polycystic Ovary Syndrome PCOS is the most common cause of AE in women, affecting 5–10% of women of reproductive age. According to the 2003 Rotterdam criteria, PCOS is diagnosed when two of the following three features are present: ultrasound appearance of polycystic ovarian morphology (PCOM), anovulation (AO), and AE. PCOS is also a major metabolic disorder, associated with IR, visceral adiposity and obesity, dyslipidemia, NAFLD, and CV disease. PCOS-associated metabolic dysfunctions intimately linked with AE; conventionally, circulating androgen burden has been typically evaluated by measuring serum testosterone (T), but recent work has defined androstenedione (A4) as a more sensitive marker for detecting PCOS-related AE and has demonstrated that integrated assessment of A4 and T is predictive of adverse metabolic risk (O’Reilly et al. 2014). Women with Monogenic Causes of Androgen Excess The variants of congenital adrenal hyperplasia (CAH) represent a group of inborn disorders with autosomal recessive inheritance characterized by glucocorticoid deficiency and variable impact on mineralocorticoid and androgen secretion. In affected women, there are three CAH variants AE-associated: 21-hydroxylase deficiency, 11β-hydroxylase deficiency, and 3β-hydroxysteroid dehydrogenase type 2 deficiency. The most common of these defects is 21-hydroxylase deficiency and results in a nonclassic CAH form with only mild glucocorticoid deficiency but relevant AE (Speiser and White 2003). As a consequence of the enzymatic block, precursor steroids are shunted down the pathways of androgen biosynthesis, which is further increased by enhanced hypothalamic-pituitary-adrenal drive due to the loss of the negative feedback by cortisol. While patients with major loss-of-function mutations usually present at birth or in early childhood, patients with mild mutations are often only diagnosed in early adulthood, as their glucocorticoid and mineralocorticoid secretion is sufficiently upheld by continuously increased ACTH stimulation of the adrenals, at the expense of AE. These patients usually do not present with outright virilization but generally with a PCOS phenotype in adolescence or early adulthood, including hirsutism, irregular periods, and PCO appearance of the ovaries. In patients with nonclassic CAH, an increased prevalence of obesity and insulin resistance has been reported, mirroring the adverse metabolic phenotype found in PCOS. As PCOS represents a diagnosis of exclusion and on average 2–3% of women presenting with a PCOS phenotype are identified as suffering from nonclassic CAH, screening for CAH by baseline serum 17-hydroxyprogesterone is recommended in the work-up of PCOS. Women with Monogenic Insulin Resistance Severe IR can develop independently of obesity as a consequence of monogenic gene defects impacting on insulin signaling or adipose tissue development. Defects in insulin signaling can be found at the level of the insulin receptor or in postreceptor signal transduction. Monogenic disorders may also cause severe obesity and consequent IR or dysfunctional adipose tissue development resulting in congenital complete or partial lipodystrophy (Semple et al. 2011). Patients with IR due to

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

7

monogenic lipodystrophy or insulin receptor (INSR) mutations present with AE, ovulatory dysfunction, PCO, and acanthosis nigricans, usually in the absence of obesity. Compensatory hyperinsulinemia may stimulate ovarian androgen biosynthesis by direct effects of insulin on theca and stromal cells, although other peripheral sources of insulin-stimulated androgen generation cannot be discounted.

Androgen Deficiency in Men and Related Metabolic Consequences Male AD is a clinical syndrome arising from failure of testicular T production, in the context of primary testicular pathology or hypothalamic-pituitary disease. In adult men, it is diagnosed by the presence of physical symptoms of AD with biochemical evidence of low circulating T. Common symptoms are a reduction of libido and erectile strength, fatigue, reduced physical strength and endurance as well as sometimes impaired cognitive function and mood disturbances (Boehm et al. 2015).

Primary Male Hypogonadism Primary male hypogonadism (HG) is defined by low serum T in combination with increased luteinizing hormone (LH). Normal T and high LH levels characterize compensated hypogonadism, which represents impaired testicular function that is rescued by increased LH stimulation. Compensated hypogonadism is subclinical but increases the likelihood to progress to overt AD when compared to the eugonadal state. Congenital primary HG can be caused by gonadal dysgenesis and cryptorchidism, as well as by autosomal or sex chromosome aneuploidies like in Klinefelter syndrome. Secondary Male Hypogonadism Secondary HG, or hypogonadotropic HG, is defined by low T and reduced gonadotrophin secretion due to impaired hypothalamic-pituitary stimulation of testicular androgen synthesis. The overwhelming majority of such cases are caused by tumors of the hypothalamic-pituitary area. Congenital hypogonadotropic hypogonadism may be observed in the context of multiple pituitary hormone deficiencies in conditions such as septo-optic dysplasia but more commonly is associated with isolated gonadotrophin deficiency as observed in Kallmann syndrome, which may be associated with anosmia and craniofacial abnormalities. Acquired Male Hypogonadism Acquired HG may be caused by lesions or tumors of the central nervous system or testis, radio- and chemotherapy, pharmacological treatment, chronic illness, poor health, and obesity. Surgical or pharmacological androgen deprivation therapy is an established treatment option for both metastatic hormone-naive and castrationresistant prostate cancer. Aging affects the hypothalamic-pituitary-gonadal (HPG) axis and can lead to lateonset AD, which is defined as low T levels if any form of classical causes of AD can be excluded. Aging can result in gradual development of testicular failure due to a

8

M. Perticone and F. Perticone

decreased number and response to LH of Leydig cells and in reduced hypothalamicpituitary signaling. Male AD can also be induced by obesity (Kelly and Jones 2015). Obesity significantly increases the age-related T decline and is associated with disordered gonadotrophin release. Conversely, weight loss can reverse obesity-associated hypogonadism. The concept of a hypogonadal-obesity-adipokine cycle is a proposed mechanism behind this association. Obesity has been suggested to lead to enhanced aromatization of androgens to estrogens by aromatase in adipose tissue, thereby reducing the level of active androgens. Estrogens may suppress the HPG axis, which reduces gonadal T synthesis.

The Role of Androgens in Metabolic Target Tissues In addition to their central role in the development and maintenance of male and female reproduction and sex drive, androgens exert key effects on metabolic target tissues. These include adipose tissue and skeletal muscle, compartments crucially involved in maintaining systemic glucose and lipid homeostasis.

Androgens, Adipose Tissue, and Lipid Metabolism Patterns of body fat distribution show a clear sexual dimorphism, with women showing a higher percentage of body fat than men and, on the contrary, with men having a greater total lean mass. The typical fat distribution in women is in a gynecoid manner, with less visceral but more subcutaneous fat; men have a predominant android fat distribution, with more visceral and less subcutaneous adipose tissue. Adipose tissue expansion is a consequence of both hyperplasia (adipogenesis), which is driven by proliferation of preadipocytes and their differentiation into adipocytes, and hypertrophy, which is driven by accumulation of lipid in differentiated adipocytes; both processes are major determinants of metabolic dysfunction (Demerath et al. 2007). Androgens impair adipogenesis by inhibiting proliferation and differentiation of mesenchymal stem cells and preadipocytes. Dihydrotestosterone (DHT) and T have inhibitory effects on multipotent stem cell commitment to the preadipocyte lineage, as well as on adipocyte differentiation in both sexes. In addition, dehydroepiandrosterone (DHEA), but not DHEA-sulfate (DHEAS), inhibits proliferation and differentiation of human subcutaneous preadipocyte cell line and enhances basal glucose uptake. An impairment of adipocyte proliferation and differentiation may lead to adipocyte hypertrophy as a compensatory mechanism to increase adipose tissue mass, which could induce adipocyte dysfunction seen in IR, intracellular stress, and inflammation which, in turn, induces a pro-inflammatory, diabetogenic, and atherogenic serum profile. Androgens also exert direct and indirect effects on adipose insulin sensitivity. Differential effects of androgens on adipose tissue and skeletal muscle and implications for global metabolism can be summarized as follows: androgens may exert pro-lipogenic effects on adipose tissue, resulting in fat mass expansion; at

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

9

Fig. 1 Differential effects of androgens on adipose tissue and skeletal muscle and implications for global metabolism. T testosterone, DHT dihydrotestosterone, 11KT 11-keto-testosterone, 11KDHT 11-keto-dihydrotestosterone. (Reprinted from: Schiffer L, Kempegowda P, Arlt W, O’Reilly MW. Mechanisms in endocrinology: the sexually dimorphic role of androgens in human metabolic disease. Eur J Endocrinol 2017;177(3):R125-R143)

higher concentrations, as observed in the healthy male range, net anabolic effects on increasing skeletal muscle bulk predominate. However, with circulating androgen levels in the range of female androgen excess and male androgen deficiency, a loss of muscle mass and an increase in abdominal obesity drive the systemic phenotype and give rise to metabolic and CV disease (Fig. 1).

Insulin Resistance, Type 2 Diabetes Mellitus, and Androgen Status in Men and Women IR is defined as the impaired systemic metabolic response to insulin, which includes glucose uptake and metabolism, suppression of lipolysis and promotion of lipogenesis, as well as protein and glycogen synthesis. IR is accompanied by compensatory hyperinsulinemia, leading to an exaggerated insulin response in normally less responsive tissues, as well as disturbances in hepatic and adipose lipid metabolism. Frank hyperglycemia occurs after decompensation of the exaggerated pancreatic beta-cell response to systemic IR.

Female Androgen Excess and Insulin Resistance The presence of AE in PCOS is closely correlated with IR. Women with PCOS show a trend to progress from normal glucose tolerance (NGT) to impaired glucose

10

M. Perticone and F. Perticone

tolerance (IGT) and to type 2 DM, and obesity significantly increases the risk. Both obese and nonobese PCOS women with AE show a high prevalence of IGT and T2DM, but obesity deteriorates the diabetic phenotype. Conversely, T levels are significantly higher in women with T2DM; consequently, AE in women has been suggested as a risk factor for T2DM (Ding et al. 2006).

Male Androgen Deficiency and Insulin Resistance In men, T levels are positively associated with insulin sensitivity, and even in men with established diagnosis of type 2 DM, low T is independently associated with IR; on the contrary, in women higher T levels predict hyperglycemia.

Body Composition and Impact of Androgen Status in Men and Women Similar to gender-specific effects observed for androgen activity on systemic IR, there are sexually dimorphic effects of androgens on body composition.

Female Androgen Excess on Body Composition PCOS women with clinical and/or biochemical evidence of AE show a higher prevalence of obesity and an increased global adiposity than the general female population. Furthermore, studies on PCOS women with AE describe an increased lean mass correlating with serum T and A4, with a shift in fat distribution from a gynecoid to an android pattern (Kirchengast and Huber 2001). Male Androgen Deficiency and Body Composition In comparison with women, circulating androgens in men correlate inversely with body mass index (BMI) and visceral adiposity. A large amount of literature supports the association between low T and increased fat mass compared to eugonadal controls. BMI negatively correlates with total and free T, and waist circumference is negatively associated with total T in men. Although age is associated with decreased androgen levels, negative associations between T and total body fat mass, body fat percentage, waist circumference, and visceral adipose tissue are maintained after adjustment for age (Blouin et al. 2005).

Nonalcoholic Fatty Liver Disease (NAFLD) and Male and Female Androgen Status The term NAFLD covers a spectrum of hepatic injury induced by obesity and IR, in the absence of significant alcohol consumption. The NAFLD spectrum ranges from intrahepatic accumulation of triglycerides or simple steatosis to diffuse tissue inflammation or nonalcoholic steatohepatitis (NASH), with a risk of progression to advanced hepatic fibrosis and cirrhosis (Hazlehurst and Tomlinson 2013). NAFLD is a major metabolic complication and an emerging CV risk factor.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

11

Female Androgen Excess and NAFLD Prevalence rates of NAFLD in PCOS appear to be higher than those in the general female population; a recent meta-analysis found that patients with PCOS have an almost fourfold higher prevalence of NAFLD compared to controls with simple obesity, even if the putative causative mechanism underlying PCOS-related NAFLD remains to be elucidated. Male Androgen Deficiency and NAFLD The role of T in the pathogenesis of NAFLD has been investigated in several studies, all reporting an inverse association between serum T and NFLD. The initial mechanism hypothesized for this association was the increased visceral adiposity in the context of hypogonadism, but recent studies have pointed out a direct role for androgens on liver metabolism, resulting in an increased malonyl-CoA, a substrate for de novo lipogenesis (Schwingel et al. 2011). Moreover, synthetic anabolic steroid use has also been linked hepatic steatosis in men.

Cardiovascular Risk and Male and Female Androgen Status Female Androgen Excess and Cardiovascular Risk Several studies demonstrated that AE in PCOS is associated with higher total cholesterol and lower HDL levels, but does not affect triglycerides and LDL levels, as well as with higher values of markers of systemic inflammation, oxidative stress, and coagulation disorders, all contributing to increase the CV risk burden. Women with PCOS and AE also exhibit microvascular dysfunction due to impaired vasodilation. Data on long-term CVevents in PCOS are inconsistent: some studies concluded that there is no increased risk for large vessel disease, abdominal aortic plaque, myocardial infarction, or stroke, while others describe increases in the prevalence of myocardial infarction and angina and in the risk of coronary heart disease and stroke. Male Androgen Deficiency and Cardiovascular Risk In men, low T levels are associated with a proatherogenic lipid profile, while an inverse relation between T and triglycerides, total cholesterol, and LDL was described (Wu and von Eckardstein 2003). On the other hand, men with coronary artery disease present with lower T levels, and its severity is negatively correlated with T levels. Male AD is associated with a higher risk of all-cause mortality, and an inverse correlation exists between T levels and prospective mortality due to all causes, CV disease, and cancer.

Effects of Thyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors The thyroid is intricately related to the CV system, sharing a common embryological origin, and thyroid hormones exert effects virtually on every organ system, including the heart and the vasculature (Grais and Sowers 2014).

12

M. Perticone and F. Perticone

The thyroid gland secretes two main iodinated hormones, 3,5,30 -triiodothyronine (T3) and 3,5,30 50 -tetraiodothyronine (T4), also known as thyroxine. Both molecules can generate biological activity in target tissues by binding to the thyroid hormone receptors; however, T3 is considered the bioactive form of thyroid hormone that mediates peripheral effects and, therefore, of specific interest with respect to risk for CV events. The affinity of the thyroid hormone receptor is approximately tenfold higher for T3 than for T4; for this reason T4 must be converted to T3 to produce potent thyroid hormone receptor-mediated effects. Anyway, although T4 is a prohormone for T3, it can directly act through thyroid hormone receptors in a variety of tissues, such as blood vessels, exerting a proangiogenic effect. Thyroid hormones have a broad range of effects on the CV system, particularly on the heart. They influence cardiac status in three ways: (1) by direct genomic actions on cardiomyocytes, resulting in the regulation of the expression of target genes; (2) by extranuclear, nongenomic actions on the ion channels in the cardiomyocyte cell membrane; (3) and through the effects of T3 and T4 on the peripheral circulation, which determines CV hemodynamics, cardiac filling, and systolic contractility (Jabbar et al. 2017). Thyroid hormone activity in the cardiomyocyte regulates myocardial contractility and systolic function by the activation of the expression of genes encoding sodium-/potassium-transporting ATPases, myosin heavy chain-α, and sarcoplasmic/endoplasmic reticulum calcium ATPase 2 and negatively regulates the transcription of myosin heavy chain-β, leading to improved ventricular relaxation. Thyroid hormones also have a direct inotropic effect on the heart by positively regulating the gene expression of the β1-adrenergic receptor. In addition, thyroid hormones influence cardiac chronotropy through both genomic and nongenomic effects on components of the adrenergic receptor complex and on sodium, potassium, and calcium channels. The effect of thyroid hormones on cardiac chronotropy manifests as tachycardia and increased risk of atrial fibrillation in hyperthyroid states and as bradycardia and reduced cardiac contractility in hypothyroidism. Nongenomic effects of thyroid hormones on cardiomyocytes and the systemic vasculature include activation of sodium, potassium, and calcium membrane ion channels, effects on mitochondria, and involvement in signaling pathways of cardiomyocytes and vascular smooth muscle cells. Thyroid hormones activate phosphatidylinositol 3-kinase (PI3K)/serine/threonine protein kinase (AKT) signaling pathways, inducing production of endothelial nitric oxide and a subsequent reduction in the systemic vascular resistance. Other nongenomic actions of thyroid hormones include vasodilation, due to a reduction of vascular resistance by both an increased production of nitric oxide (NO) and an increased calcium reuptake within the arterioles, which leads to smooth muscle relaxation. The decrease in systemic vascular resistance induced by thyroid hormones, together with their direct inotropic effects, leads to an increase in cardiac output. The renin-angiotensin-aldosterone system also has an important role in the hemodynamic effects of thyroid hormones. The initial decrease in systemic vascular resistance induced by thyroid hormones leads to decreased perfusion in the kidneys, which increases renin and aldosterone levels. The activation of the renin-angiotensin-aldosterone axis leads to an increase in cardiac preload, which is another explanation for the increase in cardiac output induced by thyroid hormones.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

13

Thyroid dysfunction is very common, and the prevalence of both hypothyroidism and hyperthyroidism increases with age.

Overt and Subclinical Hyperthyroidism Overt thyrotoxicosis or hyperthyroidism is commonly caused by stimulation of the TSH receptor by autoantibodies (Graves’ disease) or as a result of autonomous production of thyroid hormones by thyroid nodules. The prevalence of overt hyperthyroidism in the general population is 0.5%. This condition is defined by elevated peripheral free thyroid hormone levels (T3 and/or T4) and a decreased or undetectable TSH. Hyperthyroidism may result from autoimmune disease, thyroid nodule autonomy, or exogenous thyroid hormone ingestion. Tachycardia is a common sign of overt hyperthyroidism, and 5–15% of patients with overt hyperthyroidism present with atrial fibrillation. Normalization of thyroid hormone levels leads to the reversion to normal sinus rhythm in approximately 60% of patients who have atrial fibrillation due to hyperthyroidism. Shortness of breath during minimal exertion is also commonly reported by patients with hyperthyroidism; however, the exact etiology of this symptom has not been clearly defined. Moreover, hyperdynamic circulation – characterized by increased preload and contractility, reduced systemic vascular resistance, and high heart rate, leading to a 50–300% increase in cardiac output – is common in overt hyperthyroidism. If overt hyperthyroidism is left untreated, or in those individuals with severe long-standing hyperthyroidism, this increased cardiac output can lead to symptoms and signs of heart failure as a result of left ventricular hypertrophy, arrhythmias, and an increase in cardiac preload secondary to fluid overload. Patients with hyperthyroidism, unless they received radioiodine therapy to induce overt hypothyroidism, have high longterm CV mortality (Feldman et al. 1986). Subclinical hyperthyroidism is defined by low circulating TSH levels with serum concentrations of T3 and T4 within the reference range. Subclinical hyperthyroidism can be caused by exogenous (i.e., secondary to excessive thyroid hormone replacement therapy or use of other drugs such as high-dose glucocorticoids) or endogenous (such as an underlying thyroid disease causing thyroid overactivity) factors. The prevalence in the general population of endogenous subclinical hyperthyroidism depends on age, sex, and iodine intake, with a reported prevalence of 0.6–1.8% in iodine-sufficient areas and as high as 9.8% in iodine-deficient areas (Cooper and Biondi 2012). Whether exogenous and endogenous subclinical hyperthyroidism are equivalent in terms of CV effects or the risk of CV disease is currently unclear. Some evidence shows a higher heart rate, increased frequency of atrial and ventricular premature beats, and a greater left ventricular mass compared with euthyroid individuals, defining a higher CV risk. Carotid intima-media thickness was also shown to be higher in patients with subclinical hyperthyroidism than in euthyroid individuals and patients with hypothyroidism. Moreover, little evidence exists about an increase in fibrinogen plasma levels, which in turn have been associated with an elevated risk of CV events. These risk factors would be expected

14

M. Perticone and F. Perticone

to lead to an elevated risk of CV disease in patients with subclinical hyperthyroidism; in particular, several studies demonstrated an association between endogenous subclinical hyperthyroidism and incident CV disease, atrial fibrillation, and cardiac dysfunction.

Overt and Subclinical Hypothyroidism Overt hypothyroidism is diagnosed when serum TSH is elevated (usually>10 mU/ L) and circulating free T4 is low (4.0–4.5 mU/L, but 10.0 mU/mL). The prevalence of subclinical hypothyroidism in the general, adult population is 4–20%. The most frequent cardiac alteration in individuals with subclinical hypothyroidism is diastolic dysfunction owing to impaired ventricular filling and relaxation. Moreover, subclinical hypothyroidism is also responsible of an impaired relaxation of vascular smooth muscle cells, which in turn induces an increase in systemic vascular resistance and arterial stiffness. Also NO bioavailability can be reduced in this clinical condition (Kahaly 2000).

Thyroid Hormones and Cardiovascular Risk Factors Hyperlipidemia Thyroid hormones are involved in lipid metabolism. Overt and subclinical hyperthyroidism do not adversely influence lipid parameters. By contrast, the association between overt hypothyroidism and hyperlipidemia has been known for many years, with some estimates showing a link in up to 90% of patients with overt hypothyroidism (Duntas 2002). Elevated plasma lipid levels are also evident in some patients with subclinical hypothyroidism, suggesting an increased risk of atherosclerosis in these individuals. The causes of hyperlipidemia in an underactive thyroid state are the decreased expression of hepatic LDL receptors – which reduces cholesterol clearance from the bloodstream – and the reduced activity of the cholesterol breaking down enzyme monooxygenase-α.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

15

Insulin Resistance IR is defined as a glucose homeostasis disorder involving a decreased sensitivity of muscles, adipose tissue, liver, and other body tissues to insulin, despite its normal or increased plasma concentration. IR is a recognized CV risk factor, since it leads to prediabetes/overt diabetes, obesity, arterial hypertension, and dyslipidemia. Normal glucose metabolism may be disrupted by either a deficit or an excess of thyroid hormones, leading to carbohydrate disorders (Maratou et al. 2009). Hyperthyroidism causes a significant increase in the level of tissue metabolism. In order to adapt to the greater energy loss, both baseline and insulin-stimulated rate of cellular glucose depletion increases as a result of the more intense glucose oxidation and lactic acid formation, the latter of which is subsequently used by the liver to accelerate gluconeogenesis and the production of endogenous glucose. Overt hyperthyroidism is often accompanied by abnormal glucose tolerance and IR. One possible explanation is the increased demand of insulin in hyperthyroid conditions, associated with accelerated metabolism, tissue IR, and increased insulin degradation. In thyrotoxicosis, increased glucose absorption occurs in the digestive tract thanks to a higher rate of stomach emptying and increased blood flow in the portal vein, which leads to postprandial hyperglycemia, characteristic of hyperthyroidism. Moreover, the effect of thyroid hormones on hepatocytes is antagonistic to insulin and stimulates glucose production in the liver by an increase in both gluconeogenesis and glycogenolysis. Thus, even if in subjects with hyperthyroidism the glucose uptake rate in peripheral tissues is increased, on the other hand, it has also been observed that anaerobic glucose metabolism stimulated by insulin is inhibited, as glycogenogenesis decreases due to the “redirection” of intracellular glucose to the process of glycolysis and generation of lactic acid. The lactic acid released from peripheral cells returns to the liver, where it becomes a substrate for the increased hepatic glucose production. On the other hand, overt hypothyroidism is considered a risk factor for IR. In this condition, a decrease in the intestinal glucose absorption rate occurs, along with a decrease in the adrenergic activity leading to a reduction in liver and muscle glycogenolysis, as well as a decrease in gluconeogenesis and baseline insulin secretion. However, a postprandial increase in insulin secretion against the background of generalized peripheral IR has also been observed, associated with a higher concentration of free fatty acids, reduced glucose uptake, and increased glucose oxidation. On the basis of these evidences, it is possible to affirm that thyroid hormones have a significant effect on glucose metabolism and the development of IR. In hyperthyroidism, impaired glucose tolerance may be the result of mainly hepatic IR, whereas in hypothyroidism it seems to prevail peripheral IR. Blood Pressure and Vascular Function The net effect of overt hyperthyroidism on blood pressure is variable depending on the increase in cardiac output versus the reduction in systemic vascular resistance (Danzi and Klein 2003). The relationship between subclinical hyperthyroidism and hypertension is less clear, with most observational studies suggest subclinical hyperthyroidism is not linked to hypertension.

16

M. Perticone and F. Perticone

Overt and subclinical hypothyroidism are associated with diastolic hypertension and impaired vascular function. The cause of hypertension in these patients is an increase in systemic vascular resistance, endothelial dysfunction, increased arterial stiffness, and low renin levels, most probably owing the lack of the normal vasodilatory effects of T3. Central arterial stiffening increases cardiac afterload and is an important predictor of all-cause mortality, as well as a precursor for atherosclerosis. Arterial stiffness can be evaluated by measuring pulse wave velocity that increases in both subclinical and overt hypothyroidism and can be improved with levothyroxine therapy. Endothelial dysfunction is involved in the early steps of atherosclerosis and is associated with an increased risk of CV events. Endothelial-dependent vasodilation is impaired in patients with hypothyroid states and can be improved by levothyroxine treatment. Both these conditions – arterial stiffness and endothelial dysfunction – seen in hypothyroid states could be mediated by hyperlipidemia and thyroid autoantibodies. Hyperlipidemia could exert its role on endothelial dysfunction and arterial stiffness in a direct way, being one of the most important causes of atherosclerosis. On the other hand, the presence of an autoimmune process might underlie the endothelial dysfunction in most patients with subclinical hypothyroidism (Taddei et al. 2003). Moreover, both hyperlipidemia and thyroid antibodies are thought to reduce the expression of endothelial nitric oxide synthase and, therefore, impair the capacity of the artery to vasodilate.

Thrombosis Overt and subclinical hyperthyroidism have been associated with alterations in the coagulation pathway, even if this hypothesis needs to be further investigated. In particular, it remains still uncertain if the reported risk of cerebrovascular events is caused by blood thrombogenicity or by alterations in the vascular tree (e.g., an increase in the carotid intima-media thickness or the presence of vascular plaques).

Effects of Growth Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Growth hormone (GH) is synthesized and secreted by somatotroph cells in the anterior pituitary gland. It acts directly on peripheral tissues via interaction with the GH receptor and indirectly via stimulation of insulin-like growth factor type 1 (IGF-1) synthesis. IGF-1 promotes glucose uptake and cellular protein synthesis. GH and IGF-1 regulate somatic growth, including cardiac development and function.

Growth Hormone Excess Acromegaly is characterized by high circulating GH and IGF-1 levels and is caused by a benign pituitary adenoma in more than 98% of cases. Metabolic disturbances in patients suffering from this condition seem to be similar to those in the insulin-

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

17

resistant state, i.e., hyperglycemia, hyperinsulinemia, and hypertriglyceridemia (Colao et al. 2004). Thus, it is not surprising that CV disease is the leading cause of mortality in these patients. Moreover, in acromegaly there is also an increase of CV risk factors, especially hypertension, impaired glucose tolerance/diabetes mellitus, dyslipidemia (high levels of triglycerides, low or normal levels of HDL cholesterol, presence of small and/or dense low-density lipoprotein cholesterol). Hypertension occurs in 20–50% of patients with acromegaly. Possible mechanisms include increased arterial stiffness due to hypertrophy and fibrosis of the arterial muscular tunica. Both blood pressure values and glycemic control improve with normalization of IGF-1. Acromegalic patients have less visceral and subcutaneous fat mass and increased intermuscular fat mass, which may be related to insulin resistance in this disease. On the other hand, acromegalic patients exhibit an enlargement of organs and soft tissues predisposing to obstructive sleep apnea (OSA), a CV risk factor per se. Acromegalic patients often exhibit cardiac histological abnormalities, such as myocyte hypertrophy, interstitial fibrosis, inflammatory cell infiltration, reduced capillary density, myofibril derangement, and extracellular collagen deposition. The impact of these changes on the structure and function of myocardial and valvular tissues is determined by the duration and severity of GH/IGF-1 excess. In the early stage of acromegaly, there is enhanced myocardial contractility, decreased systemic vascular resistance, increased cardiac output, and overall increased cardiac performance. In the intermediate stage, after about 5 years of active disease, there is biventricular hypertrophy, diastolic dysfunction, and impaired exertional cardiac performance. Late-stage acromegalic cardiomyopathy is characterized by systolic and diastolic dysfunction, increased myocardial mass, ventricular cavity dilatation, and increased systemic vascular resistance. Acromegalic cardiomyopathy is frequently present at diagnosis. Up to two thirds of patients with acromegaly meet echocardiographic criteria for left ventricular hypertrophy; patients with severe cardiomyopathy may progress to heart failure. Successful treatment of acromegaly halts the progression of cardiac dysfunction and reduces CV mortality. Several studies have documented cardiac rhythm abnormalities in acromegaly, including atrial and ventricular ectopic beats, paroxysmal atrial fibrillation, paroxysmal supraventricular tachycardia, sick sinus syndrome, bundle branch block, and ventricular tachycardia. Somatostatin analogs have been shown to reduce QT intervals and to improve the arrhythmic profile in acromegalic patients. Cardiac valve disease (aortic and mitral regurgitation) is frequently observed in acromegaly. GH/IGF-1 excess may lead to abnormal extracellular matrix regulation and thus to pathogenesis of myxomatous valvulopathy. Aortic and mitral valve dysfunction often persists despite treatment of hormonal excess. In patients with excess of GH, there is also the disruption of coagulation and the fibrinolytic system, in particular consistently high fibrinogen levels and elevated or unchanged levels of plasminogen activator inhibitor-1 (PAI-1). The biological effects of GH on its target organs are exerted through direct or indirect stimulation of production of insulin-like growth factor 1 (IGF-1). GH directly acts as a strong promoter in lipolytic signaling. In contrast, GH might also

18

M. Perticone and F. Perticone

promote lipid synthesis and storage by induction of IGF-1, which stimulates the insulin signaling pathway (Saltiel and Kahn 2001). IR is associated with an overactive vascular RAAS and impaired NO production, resulting in impaired vasodilation and, together with the increased reabsorption of sodium and water GH-mediated, in the appearance of arterial hypertension. In adipose tissue, GH is an important mediator of lipolysis and directly acts on hormone-sensitive lipase and enhances the responsiveness for beta-adrenergic activity, which might explain higher plasma triglycerides concentrations, observed in patients with acromegaly. Besides these metabolic effects counteracting insulin action and promoting the development of IR, GH and IGF-1 are both reported to increase mitochondrial oxidation capacity in animal models as well as in humans and therefore promote whole-body energy expenditure.

Growth Hormone Deficiency Both GH and IGF-1 have been suggested to have regulatory role for peripheral resistance, with accumulating evidence supporting the vasodilating effects of IGF-1. This action seems to be mediated through release of NO and/or other vasodilators from the endothelium. Moreover, IGF-1 may cause vasorelaxation through nonendothelium-dependent actions, possibly by increasing the activity of the Na+/K+ATPase in vascular smooth muscle cells. Nevertheless, conflicting results regarding blood pressure and peripheral resistance have been reported in the literature, with some studies reporting an increased prevalence of hypertension in GH-deficient adults and others demonstrating unchanged blood pressure values in this setting of patients. Untreated GH deficiency is associated with increased body fat and central adiposity, dyslipidemia (both low values of HDL and high values of LDL), endothelial dysfunction, and IR. Increased carotid intima-media thickness, a marker of early atherosclerotic development, has also been described in GH deficiency. GH replacement therapy can result in increased lean body mass and decreased visceral adipose tissue and may decrease total and LDL cholesterol levels. Endothelial dysfunction improves with GH replacement therapy, with increased flow-mediated dilation and reduced arterial stiffness due to improved NO bioavailability. Anyway, the effects of GH replacement therapy on CV outcomes are uncertain. Furthermore, echocardiography in patients with childhood- or adolescent-onset GH deficiency has revealed significant reductions in left ventricular posterior wall thickness and interventricular septal thickness, with resultant decrease in LV mass index and LV internal diameter. Thus, both GH excess and GH deficiency passively promote the development of CV disease. On the other hand, cardiomyocytes directly express receptors for GH and IGF-1.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

19

Stimulation of these receptors induces cardiac hypertrophy and affects cardiac contractility.

Effects of Adrenocorticotropic Hormone and Cortisol Disturbances on Cardiovascular Traditional and Nontraditional Cardiovascular Risk Factors Adrenocorticotropic hormone (ACTH) is synthesized and secreted by corticotroph cells of the anterior pituitary gland. The primary role of ACTH is to regulate adrenal cortisol secretion. Excess ACTH can be produced by pituitary corticotroph adenoma or, rarely, by an extrapituitary tumor (ectopic ACTH syndrome) such as small cell lung cancer, carcinoid tumor, or medullary thyroid cancer. This excess ACTH secretion results in hypercortisolism, or Cushing’s syndrome. Endogenous Cushing’s syndrome is caused by excessive secretion of ACTH in approximately 80% of cases, and by ACTH-independent causes in approximately 20% of cases that include cortisol secretion by unilateral adrenal adenomas, or by bilateral adrenal hyperplasia or dysplasia. Hypercortisolism is associated with hypertension, central obesity, insulin resistance, dyslipidemia, and alterations in clotting and platelet function. Hypertension is present in about 80% of adult patients with endogenous Cushing’s syndrome and results from changes in regulation of plasma volume, systemic vascular resistance, and vasodilation. Treatment of Cushing’s syndrome usually results in improvement or resolution of hypertension, although hypertension may persist in patients with long-standing hypercortisolism and/or coexisting essential hypertension. Abnormal glucose metabolism in Cushing’s syndrome results from the stimulation of hepatic gluconeogenesis and glycogenolysis. Patients with hypercortisolism may have impaired fasting glucose, impaired glucose tolerance, hyperinsulinemia, insulin resistance, and/or diabetes mellitus. Cushing’s syndrome has been associated with increased lipoprotein (a), decreased HDL cholesterol, and increased triglycerides. The duration of cortisol excess correlates with the degree of dyslipidemia seen. Cortisol also increases the synthesis of several coagulation factors, stimulating endothelial production of von Willebrand factor and concomitantly increasing factor VIII. Hypercortisolism may also enhance platelet aggregation and reduce plasma fibrinolytic capacity (Newell-Price et al. 2006). Cushing’s syndrome has been also associated with left ventricular hypertrophy, concentric remodeling, diastolic dysfunction, and subclinical left ventricular systolic dysfunction. Echocardiography has revealed increased interventricular septum thickness and posterior wall thickness, increased left ventricular mass index, and increased relative wall thickness in Cushing’s patients. Diastolic dysfunction has been demonstrated, with impaired early left ventricular relaxation, longer isovolumetric relaxation times, and evidence of global myocardial relaxation impairment. The abnormalities of left ventricular structure and function may be reversible with normalization of hypercortisolism.

20

M. Perticone and F. Perticone

Effects of Aldosterone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Aldosterone is a mineralocorticoid hormone produced in the adrenal gland. Aldosterone secretion is regulated primarily by the renin-angiotensin system, although other regulatory factors include serum sodium and potassium levels and ACTH. Mineralocorticoid hormones work to maintain normal sodium and potassium concentrations and to maintain normal volume status. Increasing evidence reveals that the renin-angiotensin-aldosterone system (RAAS) is inextricably involved in linking obesity, dyslipidemia, IR, CKD, and hypertension, as well as in the pathogenesis of metabolic syndrome (Sowers et al. 2009). It has been recently demonstrated that elevated plasma aldosterone levels directly contribute to IR, endothelial dysfunction, glomerular hyperfiltration, and excess glomerular and tubular leakage of albumin, leading to maladaptive CV and renal remodeling. Furthermore, it is increasingly recognized that patients with resistant hypertension tend to be overweight and often show elevated plasma and urine levels of aldosterone. Aldosterone exerts its genomic effects through mineralocorticoid receptors (MRs) binding; on the other hand, aldosterone exerts rapid, nongenomic effects that mediate maladaptive tissue remodeling throughout the CV and central nervous system, further perpetuating the metabolic syndrome, IR, and the hypertensive state.

Aldosterone and Insulin Resistance Aldosterone secretion from the adrenal gland has been classically considered to be regulated by RAAS activation in response to intravascular volume contraction. When this axis is perturbed, as seen in several clinical conditions, including metabolic syndrome, heart failure, and CKD, inappropriate aldosterone secretion occurs despite high salt and volume retention and contributes to a state of hyperaldosteronism (Whaley-Connell et al. 2010). The increased nongenomic MR signaling, in response to these elevated levels of aldosterone, is involved in the pathophysiology of IR and other components of the metabolic syndrome. In fact, the MR has a high affinity for both aldosterone and 11-beta-hydroxyglucocorticoids, the levels of which are often elevated in clinical states characterized by central obesity, a typical feature of the metabolic syndrome. Since in metabolic syndrome circulating glucocorticoid levels may be several orders of magnitude greater than aldosterone, this clinical condition is plausible that 11-beta-hydroxyglucocorticoids bind to the MR instead of aldosterone, impacting insulin metabolic signaling. It has been demonstrated that adipose tissue produces a lipid soluble factor that stimulates aldosterone secretion. Moreover, both aldosterone and glucocorticoids can interact via MRs to promote adipogenesis and increases in fat macrophage infiltration. Thus, the interaction of fat, the adrenal cortex, and aldosterone/glucocorticoids promotes further adipogenesis and inflammation in fat tissue. This means that in clinical conditions characterized by increased obesity, MR activation by glucocorticoids, in addition to aldosterone, further potentiates inflammation, oxidative stress, fibrosis, and IR (Fallo et al. 2006).

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

21

Primary hyperaldosteronism is a group of conditions in which aldosterone production is inappropriately high, resulting in the suppression of the RAAS. Hypertension is the clinical hallmark of primary hyperaldosteronism, showing a prevalence of 0.5–4.8% in patients with essential hypertension. Potassium depletion is also characteristic of hyperaldosteronism. Common causes of primary hyperaldosteronism include unilateral autonomous adrenal adenoma and unilateral or bilateral adrenal hyperplasia. A rare cause of primary hyperaldosteronism is a heritable condition known as glucocorticoid-remediable aldosteronism (GRA).

Aldosterone and Endothelial Function Endothelial dysfunction is commonly present in concert with IR and other metabolic alterations. Several vascular metabolic abnormalities have been documented in obese, insulin resistant subjects. These abnormalities include impaired insulinstimulated glucose uptake and reduced bioavailable NO. In this context, insulindependent glucose utilization is partly dependent on insulin-mediated increases in blood flow and substrate delivery to tissues. In IR, there is decreased insulin stimulation of NO bioactivity, diminished vasodilation, and impaired substrate delivery. Increasing evidence demonstrates that elevated plasma levels of aldosterone contribute to this decrease in insulin metabolic signaling in vascular tissue. Insulin-resistant individuals with obesity and elevated plasma levels of aldosterone are more prone to endothelial dysfunction because increases in RAAS generation of ROS activate redox-sensitive serine kinases, which promote serine phosphorylation of insulin receptor-1 (IRS-1) levels which, in turn, reduce engagement with phosphoinositol 3-kinase (PI3-K), with resulting diminution of protein kinase B (Akt) and atypical protein kinase activation of eNOS phosphorylation/activation.

Aldosterone and Hypertension Elevated plasma aldosterone levels are reported in hypertensive patients and have been correlated with increased left ventricular mass as well as established as a risk factor for developing hypertension. Primary aldosteronism, resulting from bilateral adrenal hyperplasia or an aldosterone-producing adenoma, occurs with prevalence estimated at 0.5% to 4.8% of the population with general hypertension and 4.5% to 22% of those with resistant hypertension. Elevated levels of aldosterone, in association with obesity and IR, promote nongenomic inflammation and oxidative stress pathways that advance the development of resistant hypertension. Beyond its ability to inhibit endothelium-dependent relaxation by decreasing NO bioavailability, aldosterone-induced perivascular fibrosis reduces vascular compliance and increases vascular stiffness, while increased Na+/H+ exchange promotes vascular smooth muscle cell proliferation. These actions potentiate the elevation of blood pressure that occurs from the classical effects of aldosterone to promote salt retention and volume expansion, causing severe hypertension.

22

M. Perticone and F. Perticone

Mechanisms contributing to hyperaldosteronism-mediated hypertension include plasma volume expansion from sodium and fluid retention and vasoconstriction from potassium depletion. Aldosterone has been shown to decrease NO bioavailability, inhibiting endothelium-dependent relaxation. Aldosterone-mediated perivascular fibrosis reduces vascular compliance.

Aldosterone and Cardiac Structure Hyperaldosteronism causes maladaptive cardiac remodeling and has been associated with left ventricular hypertrophy (LVH), cardiac fibrosis, and diastolic dysfunction. The degree of LVH seen in primary hyperaldosteronism exceeds the effects of hypertension alone. Aldosterone has also been shown to promote collagen deposition, activate inflammatory cells, and stimulate fibroblast proliferation.

Aldosterone and Congestive Heart Failure In conditions such as heart failure and myocardial infarction, aldosterone levels are elevated and contribute to pathologic cardiovascular remodeling via direct effects on collagen deposition and resultant cardiovascular fibrosis. Elevated aldosterone levels also promote endothelial dysfunction and vascular inflammation. The addition of an aldosterone antagonist is recommended in selected patients with moderately severe to severe symptoms of heart failure and reduced LVEF, or with LV dysfunction early after myocardial infarction.

Pheochromocytoma Pheochromocytomas are catecholamine-producing tumors that originate from chromaffin cells of the adrenal medulla and the sympathetic ganglia (catecholaminesecreting paragangliomas, or extra-adrenal pheochromocytomas). Patients may present asymptomatically if diagnosed after detection by adrenal imaging or genetic testing. Symptomatic patients present with hypertension (episodic or sustained) and paroxysmal symptoms such as dizziness, headache, flushing, diaphoresis, and palpitations. Hypertension is present in over 50% of patients with pheochromocytoma and may be sustained or paroxysmal. Higher variability of blood pressure has been demonstrated in pheochromocytoma compared to patients with essential hypertension and is associated with a higher incidence of target organ damage. Markers of endothelial dysfunction, such as increased carotid intima-media thickness, have been demonstrated in patients with pheochromocytoma. These changes have been attributed to the effects of excess catecholamines on vascular wall growth and thickening. Normalization of catecholamine levels after surgical removal of pheochromocytoma has been shown to improve carotid intima-media thickness and reduce carotid wall fibrosis.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

23

Excess catecholamine action can also lead to cardiomyopathy, ischemic heart disease, myocardial stunning, and, rarely, cardiogenic shock. Patients with pheochromocytoma-associated cardiomyopathy may present with pulmonary edema or with acute chest pain and myocardial ischemia/infarction. Pulmonary edema results from increased pulmonary capillary permeability, increased peripheral vascular resistance, increased hydrostatic pressure, and overfilling or constriction of efferent pulmonary veins. Myocardial ischemia or infarction may result from coronary vasospasm, with catecholamine action leading to vasoconstriction, decreased coronary blood flow, and increased cardiac oxygen demand. Catecholamine-induced cardiomyopathy has been shown to improve after surgical treatment of pheochromocytoma. Reversal of cardiomyopathy depends on early identification and treatment (Lenders et al. 2005).

Effects of Parathyroid Hormone Disturbances on Traditional and Nontraditional Cardiovascular Risk Factors Hyperparathyroidism Parathyroid hormone (PTH) plays a critical role in maintaining an adequate calciumphosphorus homeostasis. PTH affects three principal target organs to maintain calcium balance: bone, intestinal mucosa, and kidney. Hyperparathyroidism is characterized by inappropriately high levels of PTH in the setting of elevated calcium concentrations. Causes of hyperparathyroidism include an autonomous adenoma or parathyroid gland hyperplasia (primary hyperparathyroidism, PHPT) and secondary hyperparathyroidism due to chronic kidney disease or long-standing vitamin D deficiency. PHPT is a common endocrine disease which is now usually diagnosed at an asymptomatic stage. It is characterized by elevated serum calcium associated with elevated or non-suppressed levels of PTH. Renal stones, osteoporosis, and symptoms related to hypercalcemia are well-known complications, while controversy exists regarding the CV involvement in PHPT. The increased CV risk seen in patients with mild-to-moderate PHPT seems to be mediated by different mechanisms: (1) the effect of PTH on intracellular calcium that, in turn, acts on insulin sensitivity; (2) the direct effect of PTH on vascular and cardiac muscle proliferation; or (3) the dysfunction of the renin-angiotensin-aldosterone system. For these reasons, the CV risk associated with PHPT is attributable in large part to an increased prevalence of hypertension, obesity, glucose intolerance, and IR. Furthermore, vitamin D deficiency secondary to PHP has also been implicated in the increased CV risk of these patients. Among CV alterations, increased arterial stiffness and carotid intima-media thickness, endothelial dysfunction, hypertension, left ventricular hypertrophy, and diastolic dysfunction have been reported. Among metabolic disorders, impaired insulin sensitivity, high prevalence of T2DM, dyslipidemia, hyperuricemia, increased body weight, and metabolic syndrome have been shown (Procopio et al. 2014).

24

M. Perticone and F. Perticone

PTH has been largely recognized as a hormone with vascular and CV properties and paracrine or autocrine roles in the heart. PTH exerts a direct action on cardiac myocytes by activating protein kinase C leading to hypertrophic growth. This hormone is also involved in the expression of endothelial pro-atherosclerotic and pro-inflammatory parameters such as receptor advanced glycation end products and interleukin 6. Furthermore, there are evidences that PTH modulates endothelial function by increasing the production of endothelial nitric oxide synthase and its activity.

Hyperparathyroidism and Hypertension PHPT has been associated with an increased risk of hypertension, with a prevalence ranging from 40% to 65%. The exact mechanism linking PHPT to hypertension has not yet been completely elucidated. Potential explanations include altered reninangiotensin-aldosterone axis, dysfunction or structural changes in the resistance of vessels documented by altered vasodilatory response, and/or enhanced vascular constriction in response to pressor hormones. Hyperparathyroidism Glucose Metabolism The prevalence of type 2 DM in PHP has been estimated to be approximately 8%, while the prevalence of PHP in patients with type 2 DM is about 1%. The exact mechanism underlying the association between PHP and glucose metabolism disorders is still unclear. In the general population, alteration of serum calcium homeostasis is significantly correlated with the abnormality of glucose level, IR, and betacell function; in particular, calcium influences the affinity of insulin receptor and sensitivity to insulin, and PTH concentration is also an independent determinant of insulin sensitivity. Hyperparathyroidism and Atherosclerosis High levels of PTH have been related to atherogenesis in the general population possibly via vascular calcification and remodeling, through direct PTH receptor interaction on the vessels as well as indirectly via inflammation and vascular dysfunction. In patients with PHP, flow-mediated vasodilation of brachial artery – a marker of endothelial dysfunction – resulted in impaired compared to controls. Furthermore, patients with PHP show increased aortic stiffness.

Hypoparathyroidism Hypoparathyroidism is characterized by inappropriately low or undetectable PTH levels in the setting of hypocalcemia. Hypoparathyroidism may be congenital or acquired, with the surgical removal or damage to the parathyroid glands being the most common acquired cause. The signs and symptoms of this disease derive from hypocalcemia. Mild hypocalcemia may present with neuromuscular irritability such as perioral numbness, muscle cramping, paresthesias, and positive Chvostek’s and Trousseau’s signs. Severe hypocalcemia may present with carpopedal spasm, laryngospasm, tetany, and seizures.

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

25

There are case reports of decreased myocardial performance, dilated cardiomyopathy, and congestive heart failure in patients with acute and chronic hypocalcemia. The mechanisms underlying myocardial dysfunction are unclear but may be related to impaired excitation-contraction coupling.

Effects of Intestinal Dysbacteriosis on Traditional and Nontraditional Cardiovascular Risk Factors Many microbes inhabit the human gastrointestinal tract including viruses and bacteria, fungi and protists, which make up the intestines’ symbiotic microbes. The number of bacteria carried by the human body is approximate to 1014, mostly including anaerobes; the diversity and richness of species are variable between individuals. The intestinal microbiota has many crucial functions in human health and can be considered as a virtual organ with endocrine function. It is directly involved in the body’s nutrient absorption, growth and development, biological barriers, immune regulation, metabolism, and many other aspects (Cani and Delzenne 2007). Plenty of evidence indicates that the gut microbiota is closely related to the most important CV risk factors.

Gut Microbiota Disturbances and Hyperlipidemia Recent studies demonstrate that the gut microbiota can explain 6.0% of the variation in triglycerides and 4.0% of that in HDL-c and 4.5% of that in BMI, independent of age, sex, and genetics in the general population. Furthermore, it has been demonstrated that individuals with low microbial richness have increased fasting triglycerides and decreased HDL-c. Reverse cholesterol transport (RCT) is a key pathway involving the return of excess cholesterol from peripheral tissues to the liver to excretion of bile and eventually feces. Even if the exact mechanisms of action have not been still fully elucidated, it is possible to affirm that intestinal flora metabolites are closely related to lipid metabolism (Fu et al. 2015).

Gut Microbiota Disturbances and Obesity Recent studies indicate that the increase in the number of Firmicutes and the reduction of Bacteroides in the gut microbiota are associated with obesity. Enterobacter cloacae B29 – internationally recognized as the first “fat bacteria” – has been demonstrated to be a direct cause of obesity. The gut microbiota may cause obesity by reducing fasting-induced adipokine factor (FIAF) expression. Intestinal flora can also stimulate the production of various

26

M. Perticone and F. Perticone

inflammatory factors leading to chronic systemic inflammation and further cause obesity and insulin resistance (Kvit and Kharchenko 2017). Obesity is often a leading cause for the development of T2DM, so the abovementioned mechanisms of intestinal flora causing obesity can also increase the risk of T2DM (Larsen et al. 2010).

Gut Microbiota Disturbances and Atherosclerosis Hyperlipidemia is an independent risk factor for atherosclerosis. We have previously discussed the effects of gut microbiota on lipid metabolism, which consequently affect the development of atherosclerosis. Furthermore, some studies also suggest that the GM is directly related to the occurrence of atherosclerosis, even if the core mechanism that causes atherosclerosis remains still unclear (Koren et al. 2011).

Gut Microbiota Disturbances and Cardiovascular Disease The mechanism of intestinal flora affecting cardiovascular disease mainly includes three aspects: (a) intestinal flora disorder leads to bacterial endotoxin translocation and promotes the release of inflammatory factors leading to an inflammatory response; (b) intestinal flora disorder leads to abnormal metabolism of substances, which causes CVD such as lipid, glucose, and tryptophan metabolism; (c) intestinal flora disorder promotes oxidative stress in the body and aggravates the development of CVD. Gut microbiota is also involved in the metabolism of purine and uric acid. For example, xanthine dehydrogenase, the key enzyme responsible for the oxidative metabolism of purines, is generated by the secretion of Escherichia coli in intestinal bacteria. Therefore, the decomposing activity of gut microbiota on uric acid is positively related to the content of Escherichia coli. Increased blood uric acid levels can lead to high level nitrite/nitrate in the blood, decreased bioavailability of NO, and oxidative stress.

Summary The risk of cardiovascular diseases, both in terms of morbidity and mortality, can increase in the general population because of a number of factors ranging from gene polymorphisms to modifiable environmental factors. The knowledge of major pathogenic mechanisms influencing cardiovascular anatomy, homeostasis, and function can allow a pharmacologic modulation of traditional risk factors including unbalanced diet, lifestyle, hypertension, dyslipidemia, smoking, obesity, etc. On the other hand, innovative therapeutic approaches and primary prevention strategies originate from an adequate management of nontraditional risk factors as low-grade chronic inflammation, homocysteine, environment-induced oxidative stress and/or endothelial dysfunction, lipoprotein homeostasis, psychosocial factors, insulin levels, uric acid, chronic kidney disease, and activation of the renin-angiotensin

1

Impact of Endocrine Disorders on Typical and Atypical Cardiovascular. . .

27

system. Considering the complex picture of cardiovascular effects deriving from disorders of the endocrine system, a normal endocrine function is essential for overall cardiovascular health. Disorders of the endocrine system do have multiple effects on the cardiovascular system, in particular when hyper- or hypofunction develop.

Cross-References ▶ Impact of Endocrine Disorders on Blood Pressure ▶ Impact of Endocrine Disorders on the Heart ▶ Impact of Endocrine Disorders on Vasculature ▶ Impact of Endocrine Disorders on the Kidney

References Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr Rev. 2008;29:76–131. Blouin K, Després JP, Couillard C, et al. Contribution of age and declining androgen levels to features of the metabolic syndrome in men. Metabolism. 2005;54:1034–40. Boehm U, Bouloux PM, Dattani MT, et al. Expert consensus statement on congenital hypogonadotropic hypogonadism – pathogenesis, diagnosis and treatment. Nat Rev Endocrinol. 2015;11(9):547–64. Cani PD, Delzenne NM. Gut microflora as a target for energy and metabolic homeostasis. Curr Opin Clin Nutr Metab Care. 2007;10:729–34. Colao A, Ferone D, Marzullo P, Lombardi G. Systemic complications of acromegaly: epidemiology, pathogenesis, and management. Endocr Rev. 2004;25:102–52. Cooper DS, Biondi B. Subclinical thyroid disease. Lancet. 2012;379:1142–54. Danzi S, Klein I. Thyroid hormone and blood pressure regulation. Curr Hypertens Rep. 2003;5:513–20. Dawber TR, Meadors GF, Moore FE Jr. Epidemiological approaches to heart disease: the Framingham study. Am J Public Health. 1951;41:279–86. Demerath EW, Sun SS, Rogers N, et al. Anatomical patterning of visceral adipose tissue: race, sex, and age variation. Obesity. 2007;15:2984–93. Ding EL, Song Y, Malik VS, Liu S. Sex differences of endogenous sex hormones and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA. 2006;295:1288–99. Duntas LH. Thyroid disease and lipids. Thyroid. 2002;12:287–93. Fallo F, Veglio F, Bertello C, et al. Prevalence and characteristics of the metabolic syndrome in primary hyperaldosteronism. J Clin Endocrinol Metab. 2006;91:454–9. Feldman T, Borow K, Sarne DH, Neumann A, Lang RM. Myocardial mechanics in hyperthyroidism: importance of left ventricular loading conditions, heart rate and contractile state. J Am Coll Cardiol. 1986;7:967–74. Fu J, Bonder MJ, Cenit MC, et al. The gut microbiome contributes to a substantial proportion of the variation of blood lipids. Circ Res. 2015;117:817–24. Grais IM, Sowers JR. Thyroid and the heart. Am J Med. 2014;127:691–8. Haffner SM, Lehto S, Ronnemaa T. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med. 1998;339:229–34. Hazlehurst JM, Tomlinson JW. Non-alcoholic fatty liver disease in common endocrine disorders. Eur J Endocrinol. 2013;169:R27–37. Jabbar A, Pingitore A, Pearce SHS, Zaman A, Iervasi G, Razvi S. Thyroid hormones and cardiovascular disease. Nat Rev Cardiol. 2017;14:39–55.

28

M. Perticone and F. Perticone

Kahaly GJ. Cardiovascular and atherogenic aspects of subclinical hypothyroidism. Thyroid. 2000;10:665–79. Kelly DM, Jones TH. Testosterone and obesity. Obes Rev. 2015;16:581–606. Kirchengast S, Huber J. Body composition characteristics and body fat distribution in lean women with polycystic ovary syndrome. Hum Reprod. 2001;16:1255–60. Koren O, Spor A, Felin J, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci U S A. 2011;108:4592–8. Kvit KB, Kharchenko NV. Gut microbiota changes as a risk factor for obesity. Wiad Lek. 2017;70:231–5. Larsen N, Vogensen FK, van den Berg FW, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085. Lenders JW, Eisenhofer G, Mannelli M, Pacak K. Phaeochromocytoma. Lancet. 2005;366:665–75. Maratou E, Hadjidakis DJ, Kollias A, et al. Studies of insulin resistance in patients with clinical and subclinical hypothyroidism. Eur J Endocrinol. 2009;160:785–90. Newell-Price J, Bertagna X, Grossman AB, Nieman LK. Cushing’s syndrome. Lancet. 2006;367:1605–17. O’Reilly MW, Taylor AE, Crabtree NJ, et al. Hyperandrogenemia predicts metabolic phenotype in polycystic ovary syndrome: the utility of serum androstenedione. J Clin Endocrinol Metab. 2014;99:1027–36. Procopio M, Barale M, Bertaina S, et al. Cardiovascular risk and metabolic syndrome in primary hyperparathyroidism and their correlation to different clinical forms. Endocrine. 2014;47:581–9. Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 2001;414:799–806. Schwingel PA, Cotrim HP, Salles BR, et al. Anabolic-androgenic steroids: a possible new risk factor of toxicant-associated fatty liver disease. Liver Int. 2011;31:348–53. Semple RK, Savage DB, Cochran EK, Gorden P, O’Rahilly S. Genetic syndromes of severe insulin resistance. Endocr Rev. 2011;32:498–514. Sowers JR, Whaley-Connell A, Epstein M. The emerging clinical implications of the role of aldosterone in the metabolic syndrome and resistant hypertension. Ann Intern Med. 2009;150:776–83. Speiser PW, White PC. Congenital adrenal hyperplasia. N Engl J Med. 2003;349:776–88. Taddei S, et al. Impaired endohelium-dependent vasodilation in subclinical hypothyroidism: beneficial effect of levothyroxine therapy. J Clin Endocrinol Metab. 2003;7:411–4. Valassi E, Klibanski A, Biller BM. Potential cardiac valve effects of dopamine agonists in hyperprolactinemia. J Clin Endocrinol Metab. 2010;95:1025–33. Whaley-Connell A, Johnson MS, Sowers JR. Aldosterone: role in the cardiometabolic syndrome and resistant hypertension. Progr Cardiovasc Dis. 2010;52:401–9. Wu FC, von Eckardstein A. Androgens and coronary artery disease. Endocr Rev. 2003;24:183–217. Zarotsky V, Huang MY, Carman W. Systematic literature review of the risk factors, comorbidities, and consequences of hypogonadism in men. Andrology. 2014;2:819–34.

2

Impact of Endocrine Disorders on Blood Pressure Federico Carbone, Aldo Bonaventura, Alessandra Vecchiè, Matteo Casula, Luca Liberale, Franco Dallegri, and Fabrizio Montecucco

Contents Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Diabetes and Metabolic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . The Crosstalk Between Diabetes and Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Pathophysiological Interactions Between Diabetes and Hypertension . . . . . . . . . . . . . . . . . . . . . . The Effect of Antidiabetic Drugs in Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Thyroid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hyperthyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hypothyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

31 31 31 33 34 40 40 43

F. Carbone · A. Bonaventura · A. Vecchiè · M. Casula First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy e-mail: [email protected]; [email protected]; [email protected]; [email protected] L. Liberale First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland e-mail: [email protected] F. Dallegri First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy IRCCS Ospedale Policlinico San Martino – Italian Cardiovascular Network, Genoa, Italy e-mail: [email protected] F. Montecucco (*) Ospedale Policlinico San Martino, Genoa, Italy First Clinic of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), Department of Internal Medicine, University of Genoa, Genoa, Italy IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Nework, Genoa, Italy e-mail: [email protected] © Springer Nature Switzerland AG 2021 P. Portincasa et al. (eds.), Endocrinology and Systemic Diseases, Endocrinology, https://doi.org/10.1007/978-3-319-68729-2_2

29

30

F. Carbone et al.

Mineralocorticoids and Glucocorticoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Mineralocorticoid and BP Physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hypertensive Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Mineralocorticoid Hypotension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . GH and IGF-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . GH Excess (Acromegaly) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . GH Deficiency (GHD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hyperparathyroidism and Vitamin D Deficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Interactions Between Ca2+/Phosphate Homeostasis and Renin-Angiotensin-Aldosterone System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Hyperparathyroidism and BP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Vitamin D and BP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Sexual Hormones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Estrogen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Testosterone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

44 44 45 49 51 51 52 52 52 54 55 55 55 56 56 56

Abstract

Endocrine hormones contribute to the vascular tone in physiological conditions. A complex interplay between different endocrine axes orchestrates a wide range of mediators involved in blood pressure (BP) control. Endocrine disorders may then lead to a pathological rise in blood pressure, also known as secondary hypertension. Their identification may improve BP control or even properly resolve the disease, finally reducing the global cardiovascular risk burden. In addition, experimental evidence from endocrine disorders provided interesting insights about the pathophysiology of primary hypertension. In this regard, both diabetes and the calcium-/ phosphate-regulating hormones are not only considered a main cause of endocrine hypertension but also key players in development of primary hypertension. The same pathophysiological mechanisms implicated in primary hypertension are also dysregulated in case of secondary hypertension due to endocrine disorders involving thyroid hormones, mineral- and glucocorticoids, growth hormone/insulin-like growth, and sexual hormones. Therefore, secondary hypertension may represent a paradigm for understanding pathophysiological mechanism underlying the control of BP and should be considered a source to identify potential biomarkers and even therapeutic target. Here, we summarize the current knowledge linking endocrine system and the development of hypertension. Direct effects of insulin, thyroid hormones, mineral- and glucocorticoids, growth hormone/insulin-like growth factor, sexual hormones, and calcium-/phosphorus-regulating hormones will be analyzed, also considering the complex interplay between those different pathways. Both clinical evidence and interventional clinical trials will be discussed also describing the potential clinical implications. Keywords

Diabetes · Thyroid · Mineralocorticoids · Glucocorticoids · Growth hormone · Insulin-like growth factor · Vitamin D parathyroid hormone · Estrogen · Testosterone · Endothelial cell · Vascular smooth muscle cell

2

Impact of Endocrine Disorders on Blood Pressure

31

Introduction “Secondary hypertension” includes all cases of hypertensive disease for which a cause is identifiable. Among them, endocrine disorders account for about 3% of secondary forms of hypertension. Although these forms of hypertension are quite rare, the identification of the endocrine cause may lead to the improvement of blood pressure (BP) control or even to a proper resolution of the disease, finally reducing the global cardiovascular (CV) risk burden. Due to the low prevalence of endocrine disorders in hypertensive patients, the current evidence does not support screening strategies for the identification of secondary causes of hypertension. Therefore, a clinical suspicion should be raised for a potential endocrine cause of hypertension based on clinical features and presentation of these conditions. Many endocrine axes are involved in the pathophysiology of secondary hypertension. The most common causes of endocrine hypertension are an exaggerated production of mineralocorticoids (MCs), catecholamines, thyroid hormones, and glucocorticoids. Other causes of endocrine hypertension include the dysfunction of the growth factor (GH)/insulin-like growth factor (IGF)-1 axis and sexual hormones. Even though both diabetes and the alteration in calcium (Ca2+)/phosphate homeostasis are not considered a main cause of endocrine hypertension, they can contribute to the pathophysiology of hypertension. In this paragraph, we aimed at updating the current knowledge about endocrine hypertension, focusing on clinical evidence from large clinical studies and new insights in the underlying pathophysiological mechanisms.

Diabetes and Metabolic Syndrome The Crosstalk Between Diabetes and Hypertension Diabetes represents a great public health challenge with 415 million diabetic people in 2015 according to the International Diabetes Federation and a prediction of 642 million by 2040 (Zimmet et al. 2016). Nearly 70% of diabetic people aged >40 years are also affected by hypertension, especially among black and Hispanic subjects. The majority of subjects with both hypertension and glucose disorders have type 2 diabetes (T2DM) or impaired glucose tolerance. In T2DM, hypertension is typically a part of the metabolic syndrome including insulin resistance (IR), central obesity, and dyslipidemia, whereas hypertension could develop some years after the diagnosis of type 1 diabetes and manifest as diabetic nephropathy. Each pathophysiological entity can exacerbate each other. The epidemiological and pathophysiological link between diabetes and hypertension dates back to the 1930s when an increased likelihood of hypertension in diabetic subjects had been reported for the first time. The prevalence of hypertension among diabetic subjects is largely dependent on threshold and studied population. In the United Kingdom Prospective Diabetes Study, 39% of patients were hypertensive according to the criterion of BP 160/90 mmHg or of ongoing antihypertensive therapy. In

32

F. Carbone et al.

subsequent reports from large trials, the estimated prevalence was higher, increasing from 51% in 1988–1994 to 66% in 2005–2008. A long-term observational study held in Finland in 1968 stratified normotensive and nondiabetic men by tertiles of blood glucose concentration (according to 1-h post-glucose load glucose values) and found that more individuals in the upper two tertiles had hypertension 18 years later (odds ratios [OR] 1.36 and 1.71, respectively), even after adjustment for age, alcohol consumption, adiposity, and initial systolic and diastolic BP. Similar results have been found in the San Antonio Heart Study, in which 1039 nondiabetic, non-hypertensive patients were followed for 7 years; nearly 10% developed hypertension, which has been predicted by age, waist-tohip ratio, body mass index (BMI), triglyceride values, and fasting insulin. This latter, interestingly, predicted the development of hypertension both in lean and obese subjects. Overall, these findings demonstrate that people with predisposition to develop diabetes live with increased BP levels. Long before these individuals cross the diagnostic threshold for diabetes, they have been exposed to elevated BP levels for a long time. In turn, the inverse relationship with hypertension predicting T2DM was also demonstrated. In a follow-up study including 10,000 Israeli men, systolic BP has been proven as a significant predictor for T2DM irrespective of age, peripheral artery disease, and obesity. In the Atherosclerosis Risk in Communities Study, T2DM was found almost 2.5 times as likely to develop in hypertensive as in normotensive subjects. Anyway, it is important to consider that the risk for incident diabetes among hypertensive patients might be amplified by the administration of medications, such as β-blockers and thiazide/ thiazide-like diuretics, whereas results about angiotensin-converting enzyme (ACE) inhibitors are still controversial. In the Renal Insufficiency And Cardiovascular Events (RIACE) study, male sex, family history, age, and BMI have been found as independent risk factors for hypertension, with diabetes duration adding a further risk and good renal function acting as a protective agent. Hypertension and diabetes have an addictive effective on CV risk in terms of mortality and morbidity being responsible for both microvascular (retinopathy, nephropathy, peripheral artery disease, and neuropathy) and macrovascular (coronary artery disease and stroke) complications. Moreover, hypertension and diabetes together account for a 2.5- to 7.2-fold increase in CV-related relative mortality. Analyzing both Framingham cohorts, the individual CV risk for diabetic patients decreased across the past 40 years, even if the total diabetes-related CV disease burden increased. Therefore, hypertensive subjects at the time of diabetes diagnosis presented with higher rates of all-cause mortality and CV disease events. This finding has been later confirmed in diabetic patients as hypertension was associated with a 72% increase in all-cause mortality risk and a 57% in any CV disease event risk. Hence, hypertension plays a key role in the excess CV risk in diabetic patients, mainly among men, even after adjustment for other CV risk factors (Ferrannini and Cushman 2012).

2

Impact of Endocrine Disorders on Blood Pressure

33

Pathophysiological Interactions Between Diabetes and Hypertension The increase in peripheral vascular resistance and the endothelial dysfunction are main contact points between hypertension and T2DM (Fig. 1). Obesity is the main confounder between BP and T2DM and represents a risk factor for both (Balakumar et al. 2016). Obese subjects show higher levels of plasma norepinephrine than lean subjects as well as a faster urine turnover of norepinephrine in peripheral tissues and a more sustained muscle sympathetic activity. Sympathetic activity has been proven to be more pronounced in visceral than in peripheral adipose tissue. Neuroadrenergic influences have been suggested to play a role in the pathophysiological process of the metabolic syndrome and its related CV risk. Sympathetic responses reduce insulin sensitivity by hemodynamic and cellular effects and can both directly and indirectly promote the progression of organ damage. Long-term sympathetic influences in metabolic syndrome may increase BP via different mechanisms. IR can be considered an important factor, as it shares its sympatho-stimulating effect with

Diabetes

Obesity

Hypertension

Insulin resistance Insulin

Endothelial dysfunction

Glucose

Insulin receptor

ECs Promotion of Exposure of proliferation hypercoagulability Cytokines soluble GFs

β-cell dysfunction Dysfunctional

Dead

Normal blood flow

Increased blood pressure

Narrow blood vessel

Insulin

Proinsulin

Vasoconstriction SMC proliferation Adventitial hypertrophy

Fig. 1 The interplay between diabetes mellitus and hypertension. Hypertension features are increase in peripheral vascular resistance and endothelial dysfunction, whereas diabetes mellitus is characterized by insulin resistance and β-cell dysfunction. The link between those two diseases is represented by obesity, often associated with both of them and behaving as a risk factor. EC endothelial cell, GF growth factor, SMC smooth muscle cell

34

F. Carbone et al.

partial leptin resistance, high levels of free fatty acids, and low levels of ghrelin and adiponectin. In hypertensive patients, IR is marked in skeletal muscle, but exact mechanisms contributing to hypertension are not yet fully understood. While in physiological conditions insulin can cause a slight increase in blood flow by nitric oxide (NO) release and enhance acetylcholine-mediated vasodilation, in insulinresistant subjects vasodilation in response to over-physiological concentrations of insulin is lower. IR and impaired vasodilation are often found both in T2DM and hypertension, but neither the correction of endothelial dysfunction nor vasodilation has been found to improve insulin-mediated glucose uptake in hypertensive subjects. IR, alongside with a reduction in glucose transport, triggers a compensatory response by the pancreatic production of insulin resulting in hyperinsulinemia. The anabolic response to insulin may be then overstimulated in nonresistant tissues like endothelial cells (ECs), finally leading to vessel remodeling, arterial stiffness, and loss of BP autoregulation. Hyperinsulinemia may also exert a chronic anti-natriuretic renal stimulus with sodium (Na+) and water retention, which further enhances the increase of BP. This effect may also derive from the increased renal sympathetic activity and renin-angiotensin-aldosterone system (RAAS) activation, common findings in both obesity and hypertension. Glomerulosclerosis is the long-term result of this process due to persistent hyperfiltration. Another mechanism involves the baroreflex, whose activity can be impaired by obesity. The RAAS has a major role in promoting endothelial dysfunction, especially by stimulating angiotensin II (AngII) and norepinephrine release, in addition to suppress the sympatho-inhibitory effect of NO. RAAS activity is abnormally increased in obese patients, finally leading to enhanced Na+ reabsorption, increased peripheral arterial resistance, and chronic activation of sympathetic system. A role for aldosterone in obesity-related hypertension has been suggested maybe via the production of powerful mineralocorticoidreleasing factors in the adipose tissue or the capacity of oxidized particles of linoleic acid to stimulate aldosterone synthesis.

The Effect of Antidiabetic Drugs in Hypertension In past years, large evidences suggested that BP lowering in patients with hypertension and diabetes could greatly improve CV outcomes (Table 1) (1998; Curb et al. 1996; Estacio et al. 1998; Xiang et al. 2010; Hansson et al. 1998; Patel et al. 2007; Schrier et al. 2002; Tuomilehto et al. 1999). Despite this, the optimal threshold for BP reduction in patients with diabetes has been for long time a matter of debate (1998; Curb et al. 1996; Estacio et al. 1998; Xiang et al. 2010; Hansson et al. 1998; Patel et al. 2007; Schrier et al. 2002; Tuomilehto et al. 1999). Data from the UKPDS 36 trial showed that systolic BP levels 120 mmHg gave a continuous increased CV risk (Adler et al. 2000). In the UKPDS 38 study, more than 1000 patients with hypertension and T2DM were randomly assigned to two BP targets (