1
|
de Alencar AKN, Swan KF, Mahapatra S, Lindsey SH, Pridjian GC, Bayer CL. GPER Stimulation Attenuates Cardiac Dysfunction in a Rat Model of Preeclampsia. Hypertension 2024; 81:e161-e172. [PMID: 39224973 PMCID: PMC11483207 DOI: 10.1161/hypertensionaha.123.22303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Preeclampsia poses a substantial clinical challenge, characterized by maternal hypertension, cardiac dysfunction, and persistent cardiovascular risks for both the mother and offspring. Despite the known roles of the estrogen receptor (GPER [G protein-coupled estrogen receptor]) in placental development, its impact on cardiovascular aspects within a preeclampsia animal model remains unexplored. We propose that G-1, a GPER agonist, could have the potential to regulate not only hypertension but also cardiac dysfunction in rats with preeclampsia. METHODS To explore the influence of G-1 on preeclampsia, we used the reduced uterine perfusion pressure (RUPP) model. RUPP rats were administered either G-1 (100 µg/kg per day) or hydralazine (25 mg/kg per day). We conducted echocardiography to probe the intricate cardiac effects of G-1. RESULTS The RUPP rat model revealed signs of hypertension and cardiac dysfunction and alterations in gene and protein expression within placental and heart tissues. G-1 treatment reduced blood pressure and reversed cardiac dysfunction in rats with preeclampsia. In contrast, administration of the vasodilator hydralazine reduced blood pressure without an improvement in cardiac function. In addition, while G-1 treatment restored the levels of sFLT-1 (soluble fms-like tyrosine kinase-1) in RUPP rats, hydralazine did not normalize this antiangiogenic factor. CONCLUSIONS The therapeutic intervention of G-1 significantly mitigated the cardiovascular dysfunction observed in the RUPP rat model of preeclampsia. This discovery underscores the broader significance of understanding GPER's role in the context of preeclampsia-related cardiovascular complications.
Collapse
Affiliation(s)
| | - Kenneth F. Swan
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Smruti Mahapatra
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, 70112, USA
| | - Gabriella C. Pridjian
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Carolyn L. Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
2
|
Elias-Llumbet A, Sharmin R, Berg-Sorensen K, Schirhagl R, Mzyk A. The Interplay between Mechanoregulation and ROS in Heart Physiology, Disease, and Regeneration. Adv Healthc Mater 2024; 13:e2400952. [PMID: 38962858 DOI: 10.1002/adhm.202400952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/16/2024] [Indexed: 07/05/2024]
Abstract
Cardiovascular diseases are currently the most common cause of death in developed countries. Due to lifestyle and environmental factors, this problem is only expected to increase in the future. Reactive oxygen species (ROS) are a key player in the onset of cardiovascular diseases but also have important functions in healthy cardiac tissue. Here, the interplay between ROS generation and cardiac mechanical forces is shown, and the state of the art and a perspective on future directions are discussed. To this end, an overview of what is currently known regarding ROS and mechanosignaling at a subcellular level is first given. There the role of ROS in mechanosignaling as well as the interplay between both factors in specific organelles is emphasized. The consequences at a larger scale across the population of heart cells are then discussed. Subsequently, the roles of ROS in embryogenesis, pathogenesis, and aging are further discussed, exemplifying some aspects of mechanoregulation. Finally, different models that are currently in use are discussed to study the topics above.
Collapse
Affiliation(s)
- Arturo Elias-Llumbet
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
- Laboratory of Genomic of Germ Cells, Biomedical Sciences Institute, Faculty of Medicine, University of Chile, Independencia, Santiago, 1027, Chile
| | - Rokshana Sharmin
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | | | - Romana Schirhagl
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | - Aldona Mzyk
- DTU Health Tech, Ørsteds Plads Bldg 345C, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
3
|
Gharagozloo K, Mehdizadeh M, Heckman G, Rose RA, Howlett J, Howlett SE, Nattel S. Heart Failure With Preserved Ejection Fraction in the Elderly Population: Basic Mechanisms and Clinical Considerations. Can J Cardiol 2024; 40:1424-1444. [PMID: 38604339 DOI: 10.1016/j.cjca.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) refers to a clinical condition in which the signs of heart failure, such as pulmonary congestion, peripheral edema, and increased natriuretic peptide levels, are present despite normal ejection fractions and the absence of other causes (eg, pericardial disease). The ejection fraction cutoff for the definition of HFpEF has varied in the past, but recent society guidelines have settled on a consensus of 50%. HFpEF is particularly common in the elderly population. The aim of this narrative review is to summarize the available literature regarding HFpEF in elderly patients in terms of evidence for the age dependence, specific clinical features, and underlying mechanisms. In the clinical arena, we review the epidemiology, discuss distinct clinical phenotypes typically seen in elderly patients, the importance of frailty, the role of biomarkers, and the role of medical therapies (including sodium-glucose cotransport protein 2 inhibitors, renin-angiotensin-aldosterone system blockers, angiotensin receptor/neprilysin inhibitors, diuretics, and β-adrenergic receptor blockers). We then go on to discuss the basic mechanisms implicated in HFpEF, including cellular senescence, fibrosis, inflammation, mitochondrial dysfunction, enhanced production of reactive oxygen species, abnormal cellular calcium handling, changes in microRNA signalling, insulin resistance, and sex hormone changes. Finally, we review knowledge gaps and promising areas of future investigation. Improved understanding of the specific clinical manifestations of HFpEF in elderly individuals and of the fundamental mechanisms that contribute to the age-related risk of HFpEF promises to lead to novel diagnostic and treatment approaches that will improve outcomes for this common cardiac disorder in a vulnerable population.
Collapse
Affiliation(s)
- Kimia Gharagozloo
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada
| | - Mozhdeh Mehdizadeh
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada
| | - George Heckman
- Schlegel Research Institute for Aging and University of Waterloo, Waterloo, Ontario, Canada
| | - Robert A Rose
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan Howlett
- Libin Cardiovascular Institute and Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Susan E Howlett
- Departments of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Stanley Nattel
- Montreal Heart Institute Research Center and Université de Montréal, Montréal, Quebec, Canada; McGill University Departments of Pharmacology and Therapeutics, Montréal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
4
|
Abstract
Heart failure (HF) is a significant public health problem worldwide. It has long been noted that premenopausal women, compared to postmenopausal women and men, have lower rates for developing this disease, as well as subsequent morbidity and mortality. This difference has been attributed to estrogen playing a cardioprotective role in these women, though exactly how it does so remains unclear. In this review, we examine the presence of estrogen receptors within the cardiovascular system, as well as the role they play behind the cardioprotective effect attributed to estrogen. Furthermore, we highlight the underlying mechanisms behind their alleviation of HF, as well as possible treatment approaches, such as hormone replacement therapy and exercise regimens, to manipulate these mechanisms in treating and preventing HF.
Collapse
Affiliation(s)
- Chenyue Qian
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Jingjin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of GeriatricsThe Second Clinical Medical CollegeThe First Affiliated Hospital, Shenzhen People's HospitalJinan UniversitySouthern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China.
| | - Huadong Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Department of GeriatricsThe Second Clinical Medical CollegeThe First Affiliated Hospital, Shenzhen People's HospitalJinan UniversitySouthern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Alencar AKN, Swan KF, Pridjian G, Lindsey SH, Bayer CL. Connecting G protein-coupled estrogen receptor biomolecular mechanisms with the pathophysiology of preeclampsia: a review. Reprod Biol Endocrinol 2023; 21:60. [PMID: 37393260 DOI: 10.1186/s12958-023-01112-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Throughout the course of pregnancy, small maternal spiral arteries that are in contact with fetal tissue undergo structural remodeling, lose smooth muscle cells, and become less responsive to vasoconstrictors. Additionally, placental extravillous trophoblasts invade the maternal decidua to establish an interaction between the fetal placental villi with the maternal blood supply. When successful, this process enables the transport of oxygen, nutrients, and signaling molecules but an insufficiency leads to placental ischemia. In response, the placenta releases vasoactive factors that enter the maternal circulation and promote maternal cardiorenal dysfunction, a hallmark of preeclampsia (PE), the leading cause of maternal and fetal death. An underexplored mechanism in the development of PE is the impact of membrane-initiated estrogen signaling via the G protein-coupled estrogen receptor (GPER). Recent evidence indicates that GPER activation is associated with normal trophoblast invasion, placental angiogenesis/hypoxia, and regulation of uteroplacental vasodilation, and these mechanisms could explain part of the estrogen-induced control of uterine remodeling and placental development in pregnancy. CONCLUSION Although the relevance of GPER in PE remains speculative, this review provides a summary of our current understanding on how GPER stimulation regulates some of the features of normal pregnancy and a potential link between its signaling network and uteroplacental dysfunction in PE. Synthesis of this information will facilitate the development of innovative treatment options.
Collapse
Affiliation(s)
| | - Kenneth F Swan
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Gabriella Pridjian
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, 70112, USA
| | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA.
| |
Collapse
|
6
|
Prossnitz ER, Barton M. The G protein-coupled oestrogen receptor GPER in health and disease: an update. Nat Rev Endocrinol 2023:10.1038/s41574-023-00822-7. [PMID: 37193881 DOI: 10.1038/s41574-023-00822-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 05/18/2023]
Abstract
Oestrogens and their receptors contribute broadly to physiology and diseases. In premenopausal women, endogenous oestrogens protect against cardiovascular, metabolic and neurological diseases and are involved in hormone-sensitive cancers such as breast cancer. Oestrogens and oestrogen mimetics mediate their effects via the cytosolic and nuclear receptors oestrogen receptor-α (ERα) and oestrogen receptor-β (ERβ) and membrane subpopulations as well as the 7-transmembrane G protein-coupled oestrogen receptor (GPER). GPER, which dates back more than 450 million years in evolution, mediates both rapid signalling and transcriptional regulation. Oestrogen mimetics (such as phytooestrogens and xenooestrogens including endocrine disruptors) and licensed drugs such as selective oestrogen receptor modulators (SERMs) and downregulators (SERDs) also modulate oestrogen receptor activity in both health and disease. Following up on our previous Review of 2011, we herein summarize the progress made in the field of GPER research over the past decade. We will review molecular, cellular and pharmacological aspects of GPER signalling and function, its contribution to physiology, health and disease, and the potential of GPER to serve as a therapeutic target and prognostic indicator of numerous diseases. We also discuss the first clinical trial evaluating a GPER-selective drug and the opportunity of repurposing licensed drugs for the targeting of GPER in clinical medicine.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
- Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
7
|
Arterburn JB, Prossnitz ER. G Protein-Coupled Estrogen Receptor GPER: Molecular Pharmacology and Therapeutic Applications. Annu Rev Pharmacol Toxicol 2023; 63:295-320. [PMID: 36662583 PMCID: PMC10153636 DOI: 10.1146/annurev-pharmtox-031122-121944] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and β), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein-coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.
Collapse
Affiliation(s)
- Jeffrey B Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA;
| | - Eric R Prossnitz
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA;
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, and Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
8
|
Gheit REAE, Younis RL, El-Saka MH, Emam MN, Soliman NA, El-Sayed RM, Hafez YM, AbuoHashish NA, Radwan DA, Khaled HE, Kamel S, Zaitone SA, Badawi GA. Irisin improves adiposity and exercise tolerance in a rat model of postmenopausal obesity through enhancing adipo-myocyte thermogenesis. J Physiol Biochem 2022; 78:897-913. [PMID: 35996069 PMCID: PMC9684260 DOI: 10.1007/s13105-022-00915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 07/21/2022] [Indexed: 11/28/2022]
Abstract
The prevalence of obesity and its associated metabolic disorders, along with their healthcare costs, is rising exponentially. Irisin, an adipomyokine, may serve as a critical cross-organ messenger, linking skeletal muscle with adipose tissue and the liver to integrate the energy homeostasis under diet-induced obesity. We aimed to explore the putative role of irisin in the protection against obesity in a postmenopausal rat model by modulating energy expenditure (EE). Bilateral ovariectomy (OVX) was performed. After 3 weeks of recovery, the OVX rats were classified according to their dietary protocol into rats maintained on normal diets (ND) (OVX) or high-fat diet (HFD) groups. The HFD-fed animals were equally divided into OVX/HFD, or irisin-treated OVX/HFD groups. Sham rats, maintained on ND, were selected as the control group. We evaluated anthropometric, EE, and molecular biomarkers of browning and thermogenesis in inguinal white adipose tissue and skeletal muscle, and the activity of the proteins related to mitochondrial long chain fatty acid transport, oxidation, and glycolysis. HFD of OVX further deteriorated the disturbed glucose homeostasis, lipid profile, and the reduced irisin, thermogenic parameters in adipose tissue and skeletal muscle, and EE. Irisin treatment improved the lipid profile and insulin resistance. That was associated with reduced hepatic gluconeogenic enzyme activities and restored hepatic glycogen content. Irisin reduced ectopic lipid infiltration. Irisin augmented EE by activating non-shivering thermogenesis in muscle and adipose tissues and decreasing metabolic efficiency. Our experimental evidence suggests irisin's use as a potential thermogenic agent, therapeutically targeting obesity in postmenopausal patients. Irisin modulates the non-shivering thermogenesis in skeletal muscle and adipose tissue in postmenopausal model.
Collapse
Affiliation(s)
- Rehab E Abo El Gheit
- Department of Physiology, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, Egypt.
| | - Reham L Younis
- Department of Physiology, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, Egypt
| | - Mervat H El-Saka
- Department of Physiology, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, Egypt
| | - Marwa N Emam
- Department of Physiology, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, Egypt
| | - Nema A Soliman
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rehab M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, North Sinai, El-Arish, Egypt
| | - Yasser Mostafa Hafez
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Doaa A Radwan
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Howayda E Khaled
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Samar Kamel
- Physiology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Sawsan A Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, 71451, Saudi Arabia
| | - Ghada A Badawi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, North Sinai, El-Arish, Egypt
| |
Collapse
|
9
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2022; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil.,Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
10
|
Chen J, Liu Y, Pan D, Xu T, Luo Y, Wu W, Wu P, Zhu H, Li D. Estrogen inhibits endoplasmic reticulum stress and ameliorates myocardial ischemia/reperfusion injury in rats by upregulating SERCA2a. Cell Commun Signal 2022; 20:38. [PMID: 35331264 PMCID: PMC8944077 DOI: 10.1186/s12964-022-00842-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background The incidence of coronary heart disease (CHD) in premenopausal women is significantly lower than that of men of the same age, suggesting protective roles of estrogen for the cardiovascular system against CHD. This study aimed to confirm the protective effect of estrogen on myocardium during myocardial ischemia/reperfusion (MI/R) injury and explore the underlying mechanisms. Methods Neonatal rat cardiomyocytes and Sprague–Dawley rats were used in this study. Different groups were treated by bilateral ovariectomy, 17β-estradiol (E2), adenoviral infection, or siRNA transfection. The expression of sarcoplasmic reticulum Ca2+ ATPase pump (SERCA2a) and endoplasmic reticulum (ER) stress-related proteins were measured in each group to examine the effect of different E2 levels and determine the relationship between SERCA2a and ER stress. The cell apoptosis, myocardial infarction size, levels of apoptosis and serum cardiac troponin I, ejection fraction, calcium transient, and morphology changes of the myocardium and ER were examined to verify the effects of E2 on the myocardium. Results Bilateral ovariectomy resulted in reduced SERCA2a levels and more severe MI/R injury. E2 treatment increased SERCA2a expression. Both E2 treatment and exogenous SERCA2a overexpression decreased levels of ER stress-related proteins and alleviated myocardial damage. In contrast, SERCA2a knockdown exacerbated ER stress and myocardial damage. Addition of E2 after SERCA2a knockdown did not effectively inhibit ER stress or reduce myocardial injury. Conclusions Our data demonstrate that estrogen inhibits ER stress and attenuates MI/R injury by upregulating SERCA2a. These results provide a new potential target for therapeutic intervention and drug discovery in CHD. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00842-2.
Collapse
Affiliation(s)
- Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Tongda Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
11
|
Maluleke TT, Millen AME, Michel FS. The effects of estrogen deficiency and aging on myocardial deformation and motion in normotensive female rats. Menopause 2021; 29:89-95. [PMID: 34905750 DOI: 10.1097/gme.0000000000001884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Estrogen deficiency is associated with left ventricular (LV) dysfunction in postmenopausal women and ovariectomized rats. Whether the relationship between estrogen deficiency and LV dysfunction is independent of cardiovascular disease (CVD) risk factors remains uncertain. This study assessed the effects of short-term and long-term estrogen deficiency on cardiac structure and function using conventional and speckle tracking echocardiography, independent of traditional CVD risk factors. METHODS Female Sprague-Dawley rats were divided into short-term (6 wks) ovariectomized (n = 9), short-term sham-operated (n = 10), long-term (6 mo) ovariectomized (n = 8), and long-term sham-operated (n = 9) groups. Cardiac geometry, systolic and diastolic function, and myocardial deformation and motion were measured using echocardiography. RESULTS Ovariectomy had no effect on conventional echocardiography measures of cardiac structure or function. Compared with short-term, long-term groups had reduced LV internal diameter (false discovery rate [FDR] adjusted P = 0.05) and impaired relaxation (e'; FDR adjusted P = 0.0005) independent of body mass and blood pressure (BP). Global longitudinal strain was impaired in ovariectomized compared with sham-operated rats (FDR adjusted P = 0.05), but not after adjusting for body mass and BP (FDR adjusted P = 0.16). Global longitudinal strain (FDR adjusted P = 0.05), strain rate (FDR adjusted P = 0.002), and velocity (FDR adjusted P = 0.04) were impaired in long-term compared with short-term groups. Global longitudinal strain rate remained impaired after adjustments for body mass and BP (FDR adjusted P = 0.02). CONCLUSIONS Estrogen deficiency does not independently cause cardiac remodeling, LV dysfunction, or impaired myocardial deformation. Traditional CVD risk factors accompanying estrogen deficiency may account for cardiac remodeling and dysfunction observed in postmenopausal women.
Collapse
Affiliation(s)
- Tshiamo T Maluleke
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | |
Collapse
|
12
|
Zhang X, Li T, Cheng HJ, Wang H, Ferrario CM, Groban L, Cheng CP. Chronic GPR30 agonist therapy causes restoration of normal cardiac functional performance in a male mouse model of progressive heart failure: Insights into cellular mechanisms. Life Sci 2021; 285:119955. [PMID: 34520767 DOI: 10.1016/j.lfs.2021.119955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/21/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023]
Abstract
AIMS G protein-coupled estrogen receptor 30 (GPR30) activation by its agonist, G1, exhibits beneficial actions in female with heart failure (HF). Recent evidence indicates its cardiovascular benefits may also include male as well. However, whether and how GPR30 activation may limit HF progression and have a salutary role in males is unknown. We hypothesized that chronic G1 treatment improves LV and cardiomyocyte function, [Ca2+]i regulation and β-adrenergic reserve, thus limiting HF progression in male. MAIN METHODS We compared left ventricle (LV) and myocyte function, [Ca2+]i transient ([Ca2+]iT) and β-AR modulation in control male mice (12/group) and isoproterenol-induced HF (150 mg/kg s.c. for 2 days). Two weeks after isoproterenol injection, HF mice received placebo, or G1 (150 μg/kg/day s.c. mini-pump) for 2 weeks. KEY FINDINGS Isoproterenol-treated mice exhibited HF with preserved ejection fraction (HFpEF) at 2-weeks and progressed to HF with reduced EF (HFrEF) at 4-weeks, manifested by significantly increased LV time constant of relaxation (τ), decreased EF and mitral flow (dV/dtmax), which were accompanied by reduced myocyte contraction (dL/dtmax), relaxation (dR/dtmax) and [Ca2+]iT. Acute isoproterenol-superfusion caused significantly smaller increases in dL/dtmax, dR/dtmax and [Ca2+]iT. G1 treatment in HF increased basal and isoproterenol-stimulated increases in EF and LV contractility of EES. Importantly, G1 improved basal and isoproterenol-stimulated dL/dtmax, dR/dtmax and [Ca2+]iT to control levels and restored normal cardiac β-AR subtypes modulation. SIGNIFICANCE Chronic G1 treatment restores normal myocyte basal and β-AR-stimulated contraction, relaxation, and [Ca2+]iT, thereby reversing LV dysfunction and playing a rescue role in a male mouse model of HF.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Tiankai Li
- Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America; Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heng-Jie Cheng
- Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America.
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America.
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States of America.
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America.
| | - Che Ping Cheng
- Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America.
| |
Collapse
|
13
|
Sun X, Wang H, Hodge H, Wright KN, Ahmad S, Ferrario CM, Groban L. Amplifying effect of chronic lisinopril therapy on diastolic function and the angiotensin-(1-7) Axis by the G1 agonist in ovariectomized spontaneously hypertensive rats. Transl Res 2021; 235:62-76. [PMID: 33915312 DOI: 10.1016/j.trsl.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
G protein-coupled estrogen receptor (GPER) activation by G1 attenuates diastolic dysfunction from estrogen loss, which may be partly due to suppression of angiotensin II pathological actions. We aimed to determine the independent effects of 8 weeks of G1 (100 µg/kg/d, subcutaneous pellet), ACE-inhibition (ACEi; lisinopril 10 mg/kg, drinking water), or combination therapy versus vehicle in the ovariectomized (OVX) spontaneously hypertensive rat (SHR) on cardiac function and morphometrics (echocardiography), serum equilibrium of angiotensins (mass spectroscopy) and cardiac components of the RAS (Western blotting). G1 alone and when combined with ACEi enhanced myocardial relaxation (é: 30 and 17%) and diastolic wall strain (DWS: 76 and 68%) while reducing relative wall thickness (RWT: 20 and 33%) and filling pressures (E/é: 30 and 37%). Cardiac expression levels of Mas receptor (Mas-R) and ACE2 also increased in the presence of G1. Strong antihypertensive effects of lisinopril monotherapy were associated with reductions in RWT, collagen deposition and E/é without overtly altering é or DWS. Chronic ACEi also increased cardiac levels of Mas-R and AT1-R and tilted the circulating RAS toward the formation of Ang-(1-7), which was amplified in the presence of G1. In vitro studies further revealed that an inhibitor to prolyl endopeptidase (PEP), but not to neprilysin, significantly reduced serum Ang-(1-7) levels in G1-treated rats, suggesting that G1 might be increasing Ang-(1-7) formation via PEP. We conclude that activating GPER with G1 augments components of the cardiac RAS and improves diastolic function without lowering blood pressure, and that lisinopril-induced blood pressure control and cardiac alterations in OVX SHR are permissive in facilitating G1 to augment Ang-(1-7) in serum, thereby strengthening its cardioprotective benefits.
Collapse
Affiliation(s)
- Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Hunter Hodge
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Kendra N Wright
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Sarfaraz Ahmad
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina.
| |
Collapse
|
14
|
Dinh QN, Vinh A, Arumugam TV, Drummond GR, Sobey CG. G protein-coupled estrogen receptor 1: a novel target to treat cardiovascular disease in a sex-specific manner? Br J Pharmacol 2021; 178:3849-3863. [PMID: 33948934 DOI: 10.1111/bph.15521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
As an agonist of the classical nuclear receptors, estrogen receptor-α and -β (NR3A1/2), estrogen has been assumed to inhibit the development of cardiovascular disease in premenopausal women. Indeed, reduced levels of estrogen after menopause are believed to contribute to accelerated morbidity and mortality rates in women. However, estrogen replacement therapy has variable effects on cardiovascular risk in postmenopausal women, including increased serious adverse events. Interestingly, preclinical studies have shown that selective activation of the novel membrane-associated G protein-coupled estrogen receptor, GPER, can promote cardiovascular protection. These benefits are more evident in ovariectomised than intact females or in males. It is therefore possible that selective targeting of the GPER in postmenopausal women could provide cardiovascular protection with fewer adverse effects that are caused by conventional 'receptor non-specific' estrogen replacement therapy. This review describes new data regarding the merits of targeting GPER to treat cardiovascular disease with a focus on sex differences.
Collapse
Affiliation(s)
- Quynh Nhu Dinh
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Antony Vinh
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
15
|
Niță AR, Knock GA, Heads RJ. Signalling mechanisms in the cardiovascular protective effects of estrogen: With a focus on rapid/membrane signalling. Curr Res Physiol 2021; 4:103-118. [PMID: 34746830 PMCID: PMC8562205 DOI: 10.1016/j.crphys.2021.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022] Open
Abstract
In modern society, cardiovascular disease remains the biggest single threat to life, being responsible for approximately one third of worldwide deaths. Male prevalence is significantly higher than that of women until after menopause, when the prevalence of CVD increases in females until it eventually exceeds that of men. Because of the coincidence of CVD prevalence increasing after menopause, the role of estrogen in the cardiovascular system has been intensively researched during the past two decades in vitro, in vivo and in observational studies. Most of these studies suggested that endogenous estrogen confers cardiovascular protective and anti-inflammatory effects. However, clinical studies of the cardioprotective effects of hormone replacement therapies (HRT) not only failed to produce proof of protective effects, but also revealed the potential harm estrogen could cause. The "critical window of hormone therapy" hypothesis affirms that the moment of its administration is essential for positive treatment outcomes, pre-menopause (3-5 years before menopause) and immediately post menopause being thought to be the most appropriate time for intervention. Since many of the cardioprotective effects of estrogen signaling are mediated by effects on the vasculature, this review aims to discuss the effects of estrogen on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) with a focus on the role of estrogen receptors (ERα, ERβ and GPER) in triggering the more recently discovered rapid, or membrane delimited (non-genomic), signaling cascades that are vital for regulating vascular tone, preventing hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Ana-Roberta Niță
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
| | - Greg A. Knock
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Richard J. Heads
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- Cardiovascular Research Section, King’s BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s College London, UK
| |
Collapse
|
16
|
Wang J, Li HY, Shen SY, Zhang JR, Liang LF, Huang HJ, Li B, Wu GC, Zhang YQ, Yu J. The antidepressant and anxiolytic effect of GPER on translocator protein (TSPO) via protein kinase a (PKA) signaling in menopausal female rats. J Steroid Biochem Mol Biol 2021; 207:105807. [PMID: 33345973 DOI: 10.1016/j.jsbmb.2020.105807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/05/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
Postmenopausal depression is mainly caused by the deprivation of ovarian hormones during menopausal transition, it is of great importance to study on the treatment that could effectively relieve symptoms of menopausal depression with fewer side effects. Activation of G-protein-coupled estrogen receptor (GPER) has long been reported to facilitate neuronal plasticity and improve cognition in animals. Meanwhile, it could participate in regulation of intracellular signaling pathways through the characteristic of GPER, ameliorate intracellular mitochondrial function and oxidative stress. However, the impact of GPER on regulating estrogen deprived-depressant and anxious behaviors is still largely unknown. Here we used the ovariectomized female rats to imitate the condition of menopause. Owing to the lateral ventricle administration of G-1 which specifically react with GPER receptor intracerebrally, Ovariectomized (OVX) female rats showed depressive- or anxiety-like phenotypes with attenuated mitochondrial function. In addition, G-1 facilitated PKA activation, which further accelerated TSPO phosphorylation and alleviated menopausal depression- and anxiety-like behaviors. Moreover, PKA inhibitor PKI could partially antagonized the anti-anxiety and anti-depression effects of G-1. Taken together, we concluded that GPER activation might exhibit antidepressant and anxiolytic effect by elevating TSPO phosphorylation via protein kinase A signaling and rescuing the redox status in menopausal female rats.
Collapse
Affiliation(s)
- Jing Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao-Yuan Li
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jia-Rui Zhang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling-Feng Liang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hui-Jie Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bing Li
- Center Laboratories, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Gen-Cheng Wu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu-Qiu Zhang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Leptin administration during lactation leads to different nutritional, biometric, hemodynamic, and cardiac outcomes in prepubertal and adult female Wistar rats. J Dev Orig Health Dis 2021; 12:870-875. [PMID: 33517945 DOI: 10.1017/s2040174420001312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Literature reports that insults, such as hormonal disturbances, during critical periods of development may modulate organism physiology and metabolism favoring cardiovascular diseases (CVDs) later in life. Studies show that leptin administration during lactation leads to cardiovascular dysfunction in young and adult male Wistar rats. However, there are sex differences regarding CVD. Thus, the present work aimed to investigate neonatal leptin administration's consequences on different outcomes in female rats at prepubertal and adult age. Newborn Wistar female rats were divided into two groups, Leptin and Control, receiving daily subcutaneous injections of this adipokine (8 μg/100 g) or saline for the first 10 of 21 d of lactation. Nutritional, biometric, hemodynamic, and echocardiographic parameters, as well as maximal effort ergometer performance, were determined at postnatal days (PND) 30 and 150. Leptin group presented lower food intake (p = 0.0003) and higher feed efficiency (p = 0.0058) between PND 21 and 30. Differences concerning echocardiographic parameters revealed higher left ventricle internal diameter (LVID) in systole (p = 0.0051), as well as lower left ventricle ejection fraction (LVEF) (p = 0.0111) and fractional shortening (FS) (p = 0.0405) for this group at PND 30. Older rats treated with leptin during lactation presented only higher LVID in systole (p = 0.0270). Systolic blood pressure and maximum effort ergometer test performance was similar between groups at both ages. These data suggest that nutritional, biometric, and cardiac outcomes due to neonatal leptin administration in female rats are age-dependent.
Collapse
|
18
|
Abstract
PURPOSE OF THE REVIEW This review summarizes sex-related changes in the heart and vasculature that occur with aging, both in the presence and absence of cardiovascular disease (CVD). RECENT FINDINGS In the presence of CVD risk factors and/or overt CVD, sex-specific changes in the number of cardiomyocytes, extent of the myocardial extracellular matrix, and myocellular hypertrophy promote unique patterns of LV remodeling in men and women. In addition, age- and sex-specific vascular stiffening is also well established, driven by changes in endothelial dysfunction, elastin-collagen content, microvascular dysfunction, and neurohormonal signaling. Together, these changes in LV chamber geometry and morphology, coupled with heightened vascular stiffness, appear to drive both age-related increases in systolic function and declines in diastolic function, particularly in postmenopausal women. Accordingly, estrogen has been implicated as a key mediator, given its direct vasodilating properties, association with nitric oxide excretion, and involvement in myocellular Ca2+ handling, mitochondrial energy production, and oxidative stress. The culmination of the abovementioned sex-specific cardiac and vascular changes across the lifespan provides important insight into heart failure development, particularly of the preserved ejection fraction variety, while offering promise for future preventive strategies and therapeutic approaches.
Collapse
Affiliation(s)
- Andrew Oneglia
- Applied Physiology and Advanced Imaging Lab, University of Texas at Arlington, 655 West Mitchell St, Arlington, TX, 76010, USA
| | - Michael D Nelson
- Applied Physiology and Advanced Imaging Lab, University of Texas at Arlington, 655 West Mitchell St, Arlington, TX, 76010, USA
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vicente Blvd, AHSP Suite A3206, Los Angeles, CA, 90048, USA
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vicente Blvd, AHSP Suite A3206, Los Angeles, CA, 90048, USA.
| |
Collapse
|
19
|
Gohar EY. G protein-coupled estrogen receptor 1 as a novel regulator of blood pressure. Am J Physiol Renal Physiol 2020; 319:F612-F617. [PMID: 32893662 DOI: 10.1152/ajprenal.00045.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mechanisms underlying hypertension are multifaceted and incompletely understood. New evidence suggests that G protein-coupled estrogen receptor 1 (GPER1) mediates protective actions within the cardiovascular and renal systems. This mini-review focuses on recent advancements in our understanding of the vascular, renal, and cardiac GPER1-mediated mechanisms that influence blood pressure regulation. We emphasize clinical and basic evidence that suggests GPER1 as a novel target to aid therapeutic strategies for hypertension. Furthermore, we discuss current controversies and challenges facing GPER1-related research.
Collapse
Affiliation(s)
- Eman Y Gohar
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
20
|
Abstract
Purpose of Review Angiotensin-converting enzyme 2 (ACE2), a specific high-affinity angiotensin II-hydrolytic enzyme, is the vector that facilitates cellular entry of SARS-CoV-1 and the novel SARS-CoV-2 coronavirus. SARS-CoV-2, which crossed species barriers to infect humans, is highly contagious and associated with high lethality due to multi-organ failure, mostly in older patients with other co-morbidities. Recent Findings Accumulating clinical evidence demonstrates that the intensity of the infection and its complications are more prominent in men. It has been postulated that potential functional modulation of ACE2 by estrogen may explain the sex difference in morbidity and mortality. Summary We review here the evidence regarding the role of estrogenic hormones in ACE2 expression and regulation, with the intent of bringing to the forefront potential mechanisms that may explain sex differences in SARS-CoV-2 infection and COVID-19 outcomes, assist in management of COVID-19, and uncover new therapeutic strategies.
Collapse
|
21
|
Garate-Carrillo A, Gonzalez J, Ceballos G, Ramirez-Sanchez I, Villarreal F. Sex related differences in the pathogenesis of organ fibrosis. Transl Res 2020; 222:41-55. [PMID: 32289256 PMCID: PMC7721117 DOI: 10.1016/j.trsl.2020.03.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
The development of organ fibrosis has garnered rising attention as multiple diseases of increasing and/or high prevalence appear to progress to the chronic stage. Such is the case for heart, kidney, liver, and lung where diseases such as diabetes, idiopathic/autoimmune disorders, and nonalcoholic liver disease appear to notably drive the development of fibrosis. Noteworthy is that the severity of these pathologies is characteristically compounded by aging. For these reasons, research groups and drug companies have identified fibrosis as a therapeutic target for which currently, there are essentially no effective options. Although a limited body of published studies are available, most literature indicates that in multiple organs, premenopausal women are protected from developing severe forms of fibrosis suggesting an important role for sex hormones in mitigating this process. Investigators have implemented relevant animal models of organ disease linked to fibrosis supporting in general, these observations. In vitro studies and transgenic animals models have also been used in an attempt to understand the role that sex hormones and related receptors play in the development of fibrosis. However, in the setting of chronic disease in some organs such as the heart older (postmenopausal) women within a few years can quickly approach men in disease severity and develop significant degrees of fibrosis. This review summarizes the current body of relevant literature and highlights the imperative need for a major focus to be placed on understanding the manner in which sex and the presence or absence of related hormones modulates cell phenotypes so as to allow for fibrosis to develop.
Collapse
Affiliation(s)
- Alejandra Garate-Carrillo
- Department of Medicine, School of Medicine, University of California, San Diego, California; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico city, Mexico
| | - Julisa Gonzalez
- Department of Medicine, School of Medicine, University of California, San Diego, California
| | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico city, Mexico
| | - Israel Ramirez-Sanchez
- Department of Medicine, School of Medicine, University of California, San Diego, California; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico city, Mexico
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, California; VA San Diego Health Care, San Diego, California.
| |
Collapse
|
22
|
Aryan L, Younessi D, Zargari M, Banerjee S, Agopian J, Rahman S, Borna R, Ruffenach G, Umar S, Eghbali M. The Role of Estrogen Receptors in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21124314. [PMID: 32560398 PMCID: PMC7352426 DOI: 10.3390/ijms21124314] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular Diseases (CVDs) are the leading cause of death globally. More than 17 million people die worldwide from CVD per year. There is considerable evidence suggesting that estrogen modulates cardiovascular physiology and function in both health and disease, and that it could potentially serve as a cardioprotective agent. The effects of estrogen on cardiovascular function are mediated by nuclear and membrane estrogen receptors (ERs), including estrogen receptor alpha (ERα), estrogen receptor beta (ERβ), and G-protein-coupled ER (GPR30 or GPER). Receptor binding in turn confers pleiotropic effects through both genomic and non-genomic signaling to maintain cardiovascular homeostasis. Each ER has been implicated in multiple pre-clinical cardiovascular disease models. This review will discuss current reports on the underlying molecular mechanisms of the ERs in regulating vascular pathology, with a special emphasis on hypertension, pulmonary hypertension, and atherosclerosis, as well as in regulating cardiac pathology, with a particular emphasis on ischemia/reperfusion injury, heart failure with reduced ejection fraction, and heart failure with preserved ejection fraction.
Collapse
|
23
|
Manrique-Acevedo C, Chinnakotla B, Padilla J, Martinez-Lemus LA, Gozal D. Obesity and cardiovascular disease in women. Int J Obes (Lond) 2020; 44:1210-1226. [PMID: 32066824 PMCID: PMC7478041 DOI: 10.1038/s41366-020-0548-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/20/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
Abstract
As the prevalence of obesity continues to grow worldwide, the health and financial burden of obesity-related comorbidities grows too. Cardiovascular disease (CVD) is clearly associated with increased adiposity. Importantly, women are at higher risk of CVD when obese and insulin resistant, in particular at higher risk of developing heart failure with preserved ejection fraction and ischemic heart disease. Increased aldosterone and mineralocorticoid receptor activation, aberrant estrogenic signaling and elevated levels of androgens are among some of the proposed mechanisms explaining the heightened CVD risk. In addition to traditional cardiovascular risk factors, understanding nontraditional risk factors specific to women, like excess weight gain during pregnancy, preeclampsia, gestational diabetes, and menopause are central to designing personalized interventions aimed to curb the epidemic of CVD. In the present review, we examine the available evidence supporting a differential cardiovascular impact of increased adiposity in women compared with men and the proposed pathophysiological mechanisms behind these differences. We also discuss women-specific cardiovascular risk factors associated with obesity and insulin resistance.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Bhavana Chinnakotla
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Department of Child Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
24
|
Loredo-Mendoza ML, Ramirez-Sanchez I, Bustamante-Pozo MM, Ayala M, Navarrete V, Garate-Carrillo A, Ito BR, Ceballos G, Omens J, Villarreal F. The role of inflammation in driving left ventricular remodeling in a pre-HFpEF model. Exp Biol Med (Maywood) 2020; 245:748-757. [PMID: 32183553 DOI: 10.1177/1535370220912699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPACT STATEMENT The incidence of HFpEF continues to increase and ∼2/3 of the patient population are post-menopausal women. Unfortunately, most studies focus on the use of male animal models of remodeling. In this study, however, using female rats to set a model of pre-HFpEF, we provide insights to possible mechanisms that contribute to HFpEF development in humans that will lead us to a better understanding of the underlying pathophysiology of HFpEF.
Collapse
Affiliation(s)
- Maria L Loredo-Mendoza
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Histopathology, School of Medicine, Universidad Panamericana, Ciudad de Mexico 03920, Mexico
| | - Israel Ramirez-Sanchez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Moises Muratt Bustamante-Pozo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Marcos Ayala
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Viridiana Navarrete
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Alejandra Garate-Carrillo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Bruce R Ito
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Jeffrey Omens
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
- VA San Diego Healthcare, San Diego, CA 92161, USA
| |
Collapse
|
25
|
Jiao L, Machuki JO, Wu Q, Shi M, Fu L, Adekunle AO, Tao X, Xu C, Hu X, Yin Z, Sun H. Estrogen and calcium handling proteins: new discoveries and mechanisms in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2020; 318:H820-H829. [PMID: 32083972 DOI: 10.1152/ajpheart.00734.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogen deficiency is considered to be an important factor leading to cardiovascular diseases (CVDs). Indeed, the prevalence of CVDs in postmenopausal women exceeds that of premenopausal women and men of the same age. Recent research findings provide evidence that estrogen plays a pivotal role in the regulation of calcium homeostasis and therefore fine-tunes normal cardiomyocyte contraction and relaxation processes. Disruption of calcium homeostasis is closely associated with the pathological mechanism of CVDs. Thus, this paper maps out and summarizes the effects and mechanisms of estrogen on calcium handling proteins in cardiac myocytes, including L-type Ca2+ channel, the sarcoplasmic reticulum Ca2+ release channel named ryanodine receptor, sarco(endo)plasmic reticulum Ca2+-ATPase, and sodium-calcium exchanger. In so doing, we provide theoretical and experimental evidence for the successful design of estrogen-based prevention and treatment therapies for CVDs.
Collapse
Affiliation(s)
- Lijuan Jiao
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | | | - Qi Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingjin Shi
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lu Fu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | | | - Xi Tao
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenxi Xu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xide Hu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zeyuan Yin
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
26
|
G-Protein–Coupled Estrogen Receptor Agonist G1 Improves Diastolic Function and Attenuates Cardiac Renin–Angiotensin System Activation in Estrogen-Deficient Hypertensive Rats. J Cardiovasc Pharmacol 2019; 74:443-452. [DOI: 10.1097/fjc.0000000000000721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Alencar AKN, Montes GC, Costa DG, Mendes LVP, Silva AMS, Martinez ST, Trachez MM, Cunha VDMN, Montagnoli TL, Fraga AGM, Wang H, Groban L, Fraga CAM, Sudo RT, Zapata-Sudo G. Cardioprotection Induced by Activation of GPER in Ovariectomized Rats With Pulmonary Hypertension. J Gerontol A Biol Sci Med Sci 2019; 73:1158-1166. [PMID: 29790948 DOI: 10.1093/gerona/gly068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 04/26/2018] [Indexed: 01/09/2023] Open
Abstract
Pulmonary hypertension (PH) is a disease of women (female-to-male ratio 4:1), and is associated with cardiac and skeletal muscle dysfunction. Herein, the activation of a new estrogen receptor (GPER) by the agonist G1 was evaluated in oophorectomized rats with monocrotaline (MCT)-induced PH. Depletion of estrogen was induced by bilateral oophorectomy (OVX) in Wistar rats. Experimental groups included SHAM or OVX rats that received a single intraperitoneal injection of MCT (60 mg/kg) for PH induction. Animals received s.c. injection of either vehicle or G1, a GPER agonist, (400 µg/kg/day) for 14 days after the onset of disease. Rats with PH exhibited exercise intolerance and cardiopulmonary alterations, including reduced pulmonary artery flow, biventricular remodeling, and left ventricular systolic and diastolic dysfunction. The magnitude of these PH-induced changes was significantly greater in OVX versus SHAM rats. G1 treatment reversed both cardiac and skeletal muscle functional aberrations caused by PH in OVX rats. G1 reversed PH-related cardiopulmonary dysfunction and exercise intolerance in female rats, a finding that may have important implications for the ongoing clinical evaluation of new drugs for the treatment of the disease in females after the loss of endogenous estrogens.
Collapse
Affiliation(s)
- Allan K N Alencar
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Guilherme C Montes
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Daniele G Costa
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Luiza V P Mendes
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.,Departamento de Farmacologia, Universidade Estácio de Sá, Rio de Janeiro, Brazil
| | - Ananssa M S Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Sabrina T Martinez
- Departamento de Química, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Margarete M Trachez
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Valéria do M N Cunha
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Tadeu L Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Aline G M Fraga
- Faculdade de Farmácia da Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Ilha do Fundão Cidade Universitária, Brazil
| | - Hao Wang
- Departments of Anesthesiology and Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Leanne Groban
- Departments of Anesthesiology and Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Carlos A M Fraga
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Roberto T Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Wang H, Sun X, Hodge HS, Ferrario CM, Groban L. NLRP3 inhibition improves heart function in GPER knockout mice. Biochem Biophys Res Commun 2019; 514:998-1003. [PMID: 31092335 DOI: 10.1016/j.bbrc.2019.05.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
The molecular mechanisms of postmenopausal heart diseases in women may involve the loss of estrogen-deactivation of its membrane receptor, G-protein coupled estrogen receptor (GPER), and subsequent activation of the cardiac NLRP3 inflammasome, a component of the innate immune system. To study the potential effects of cardiac GPER on NLRP3-mediated inflammatory pathways, we characterized changes in innate immunity gene transcripts in hearts from 6-month-old cardiomyocyte-specific GPER knockout (KO) mice and their GPER-intact wild type (WT) littermates using RT2 Profiler™ real-time PCR array. GPER deletion in cardiomyocytes decreased %fractional shortening (%FS) and myocardial relaxation (e'), and increased the early mitral inflow filling velocity-to-early mitral annular descent velocity ratio (E/e'), determined by echocardiography, and increased the mRNA levels of atrial natriuretic factor (ANF) and brain natriuretic peptide (BNP), determined by real-time PCR. Of the 84 inflammasome-related genes tested, 9 genes were upregulated, including NLRP3 and IL-18, while 1 gene, IL-12a, was downregulated in GPER KO when compared to WT. The importance of NLRP3 upregulation in GPER KO-induced heart failure was further confirmed by an in vivo study showing that, compared to vehicle-treated KO mice, 8 weeks of treatment with a NLRP3 inhibitor, MCC950 (10 mg/kg, i.p., 3 times per week), significantly limited hypertrophic remodeling, defined by reductions in heart weight/body weight, and improved systolic and diastolic functional indices, including increases in %FS and e', and decreases E/e' (P < 0.05). Both ANF and BNP mRNA levels were also significantly reduced by chronic MCC950 treatment. The findings from this study point toward a new understanding for the increased occurrence of heart diseases in women following loss or absence of estrogenic protection and GPER activation that involves cardiac NLRP3 inflammatory pathways.
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157-1009, USA; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA.
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157-1009, USA.
| | - Hunter S Hodge
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157-1009, USA.
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA; Division of Public Health Sciences, Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA.
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157-1009, USA; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston Salem, NC, 27157, USA.
| |
Collapse
|
29
|
Yan Y, Miao D, Yang Z, Zhang D. Loss of p27 kip1 suppresses the myocardial senescence caused by estrogen deficiency. J Cell Biochem 2019; 120:13994-14003. [PMID: 30957908 DOI: 10.1002/jcb.28674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Abstract
Estrogen deficiency accelerates the aging process and increases the risk of developing cardiovascular disease (CVD). Apoptosis is one of the important mechanisms of aging. p27kip1 is a cyclin-dependent kinase inhibitor that can regulate cell cycle, apoptosis, and cell motility. p27kip1 overexpression can inhibit cell cycle and increase apoptosis so it has been considered as a marker of aging. In the present study, bilateral ovariectomy (OVX) was performed as a model for menopause in wild-type (WT) and p27kip1 knockout (KO) mice to assess the effects of p27kip1 loss in myocardial aging caused by estrogen deficiency. We found that myocardial fibrosis and heart weight/body weight ratio of mice in the OVX group and p27kip1 KO group were significantly increased. Echocardiography showed that the left ventricular diameter and volume of the WT OVX group increased significantly and the cardiac function decreased. However, there was no significant difference in the results of echocardiography between the two p27kip1 KO groups. The aging and apoptosis indexes in OVX group were increased significantly, However, the indexes in p27kip1 KO mice were decreased. The expression of antioxidant indexes in OVX group was decreased significantly and p27kip1 KO can improve the antioxidant ability. These results provided that estrogen deficiency increased oxidative stress and apoptosis, accelerated aging of heart. p27kip1 KO can partly delay the aging and apoptosis of heart through upregulated antioxidant enzymes.
Collapse
Affiliation(s)
- Yi Yan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dingguo Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Bustamante M, Garate-Carrillo A, R Ito B, Garcia R, Carson N, Ceballos G, Ramirez-Sanchez I, Omens J, Villarreal F. Unmasking of oestrogen-dependent changes in left ventricular structure and function in aged female rats: a potential model for pre-heart failure with preserved ejection fraction. J Physiol 2019; 597:1805-1817. [PMID: 30681142 DOI: 10.1113/jp277479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Heart failure with preserved ejection fraction (HFpEF) is seen more frequently in older women; risk factors include age, hypertension and excess weight. No female animal models of early stage remodelling (pre-HFpEF) have examined the effects that the convergence of such factors have on cardiac structure and function. In this study, we demonstrate that ageing can lead to the development of mild chamber remodelling, diffuse fibrosis and loss of diastolic function. The loss of oestrogens further aggravates such changes by leading to a notable drop in cardiac output (while preserving normal ejection fraction) in the presence of diffuse fibrosis that is more predominant in endocardium and is accompanied by papillary fibrosis. Excess weight did not markedly aggravate such findings. This animal model recapitulates many of the features recognized in older, female HFpEF patients and thus, may serve to examine the effects of candidate therapeutic agents. ABSTRACT Two-thirds of patients with heart failure with preserved ejection fraction (HFpEF) are older women, and risk factors include hypertension and excess weight/obesity. Pathophysiological factors that drive early disease development (before heart failure ensues) remain obscure and female animal models are lacking. The study evaluated the intersecting roles of ageing, oestrogen depletion and excess weight on altering cardiac structure/function. Female, 18-month-old, Fischer F344 rats were divided into an aged group, aged + ovariectomy (OVX) and aged + ovariectomy + 10% fructose (OVF) in drinking water (n = 8-16/group) to induce weight gain. Left ventricular (LV) structure/function was monitored by echocardiography. At 22 months of age, animals were anaesthetized and catheter-based haemodynamics evaluated, followed by histological measures of chamber morphometry and collagen density. All aged animals developed hypertension. OVF animals increased body weight. Echocardiography only detected mild chamber remodelling with ageing while intraventricular pressure-volume loop analysis showed significant (P < 0.05) decreases vs. ageing in stroke volume (13% OVX and 15% for OVF), stroke work (34% and 52%) and cardiac output (29% and 27%), and increases in relaxation time (10% OVX) with preserved ejection fraction. Histology indicated papillary and interstitial fibrosis with ageing, which was higher in the endocardium of OVX and OVF groups. With ageing, ovariectomy leads to the loss of diastolic and global LV function while preserving ejection fraction. This model recapitulates many cardiovascular features present in HFpEF patients and may help understand the roles that ageing and oestrogen depletion play in early (pre-HFpEF) disease development.
Collapse
Affiliation(s)
- Moises Bustamante
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico, DF
| | - Alejandra Garate-Carrillo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico, DF
| | - Bruce R Ito
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ricardo Garcia
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,Bristol-Myers Squibb, New York, NY, USA
| | | | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico, DF
| | - Israel Ramirez-Sanchez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico, DF
| | - Jeffrey Omens
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.,VA San Diego Health Care, San Diego, CA, USA
| |
Collapse
|
31
|
Wang J, Yu R, Han QQ, Huang HJ, Wang YL, Li HY, Wang HM, Chen XR, Ma SL, Yu J. G-1 exhibit antidepressant effect, increase of hippocampal ERs expression and improve hippocampal redox status in aged female rats. Behav Brain Res 2019; 359:845-852. [DOI: 10.1016/j.bbr.2018.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/09/2018] [Accepted: 07/20/2018] [Indexed: 01/31/2023]
|
32
|
Mahmoodzadeh S, Dworatzek E. The Role of 17β-Estradiol and Estrogen Receptors in Regulation of Ca 2+ Channels and Mitochondrial Function in Cardiomyocytes. Front Endocrinol (Lausanne) 2019; 10:310. [PMID: 31156557 PMCID: PMC6529529 DOI: 10.3389/fendo.2019.00310] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
Numerous epidemiological, clinical, and animal studies showed that cardiac function and manifestation of cardiovascular diseases (CVDs) are different between males and females. The underlying reasons for these sex differences are definitely multifactorial, but major evidence points to a causal role of the sex steroid hormone 17β-estradiol (E2) and its receptors (ER) in the physiology and pathophysiology of the heart. Interestingly, it has been shown that cardiac calcium (Ca2+) ion channels and mitochondrial function are regulated in a sex-specific manner. Accurate mitochondrial function and Ca2+ signaling are of utmost importance for adequate heart function and crucial to maintaining the cardiovascular health. Due to the highly sensitive nature of these processes in the heart, this review article highlights the current knowledge regarding sex dimorphisms in the heart implicating the importance of E2 and ERs in the regulation of cardiac mitochondrial function and Ca2+ ion channels, thus the contractility. In particular, we provide an overview of in-vitro and in-vivo studies using either E2 deficiency; ER deficiency or selective ER activation, which suggest that E2 and ERs are strongly involved in these processes. In this context, this review also discusses the divergent E2-responses resulting from the activation of different ER subtypes in these processes. Detailed understanding of the E2 and ER-mediated molecular and cellular mechanisms in the heart under physiological and pathological conditions may help to design more specifically targeted drugs for the management of CVDs in men and women.
Collapse
Affiliation(s)
- Shokoufeh Mahmoodzadeh
- Department of Molecular Muscle Physiology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Shokoufeh Mahmoodzadeh
| | - Elke Dworatzek
- Department of Molecular Muscle Physiology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Institute of Gender in Medicine, Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
33
|
Groban L, Tran QK, Ferrario CM, Sun X, Cheng CP, Kitzman DW, Wang H, Lindsey SH. Female Heart Health: Is GPER the Missing Link? Front Endocrinol (Lausanne) 2019; 10:919. [PMID: 31993020 PMCID: PMC6970950 DOI: 10.3389/fendo.2019.00919] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
The G Protein-Coupled Estrogen Receptor (GPER) is a novel membrane-bound receptor that mediates non-genomic actions of the primary female sex hormone 17β-estradiol. Studies over the past two decades have elucidated the beneficial actions of this receptor in a number of cardiometabolic diseases. This review will focus specifically on the cardiac actions of GPER, since this receptor is expressed in cardiomyocytes as well as other cells within the heart and most likely contributes to estrogen-induced cardioprotection. Studies outlining the impact of GPER on diastolic function, mitochondrial function, left ventricular stiffness, calcium dynamics, cardiac inflammation, and aortic distensibility are discussed. In addition, recent data using genetic mouse models with global or cardiomyocyte-specific GPER gene deletion are highlighted. Since estrogen loss due to menopause in combination with chronological aging contributes to unique aspects of cardiac dysfunction in women, this receptor may provide novel therapeutic effects. While clinical studies are still required to fully understand the potential for pharmacological targeting of this receptor in postmenopausal women, this review will summarize the evidence gathered thus far on its likely beneficial effects.
Collapse
Affiliation(s)
- Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
- *Correspondence: Leanne Groban
| | - Quang-Kim Tran
- Department of Physiology & Pharmacology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, United States
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Che Ping Cheng
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Dalane W. Kitzman
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
34
|
Michalson KT, Groban L, Howard TD, Shively CA, Sophonsritsuk A, Appt SE, Cline JM, Clarkson TB, Carr JJ, Kitzman DW, Register TC. Estradiol Treatment Initiated Early After Ovariectomy Regulates Myocardial Gene Expression and Inhibits Diastolic Dysfunction in Female Cynomolgus Monkeys: Potential Roles for Calcium Homeostasis and Extracellular Matrix Remodeling. J Am Heart Assoc 2018; 7:e009769. [PMID: 30571375 PMCID: PMC6404177 DOI: 10.1161/jaha.118.009769] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
Background Left ventricular ( LV ) diastolic dysfunction often precedes heart failure with preserved ejection fraction, the dominant form of heart failure in postmenopausal women. The objective of this study was to determine the effect of oral estradiol treatment initiated early after ovariectomy on LV function and myocardial gene expression in female cynomolgus macaques. Methods and Results Monkeys were ovariectomized and randomized to receive placebo (control) or oral estradiol at a human-equivalent dose of 1 mg/day for 8 months. Monkeys then underwent conventional and tissue Doppler imaging to assess cardiac function, followed by transcriptomic and histomorphometric analyses of LV myocardium. Age, body weight, blood pressure, and heart rate were similar between groups. Echocardiographic mitral early and late inflow velocities, mitral annular velocities, and mitral E deceleration slope were higher in estradiol monkeys (all P<0.05), despite similar estimated LV filling pressure. MCP1 (monocyte chemoattractant protein 1) and LV collagen staining were lower in estradiol animals ( P<0.05). Microarray analysis revealed differential myocardial expression of 40 genes (>1.2-fold change; false discovery rate, P<0.05) in estradiol animals relative to controls, which implicated pathways associated with better calcium ion homeostasis and muscle contraction and lower extracellular matrix deposition ( P<0.05). Conclusions Estradiol treatment initiated soon after ovariectomy resulted in enhanced LV diastolic function, and altered myocardial gene expression towards decreased extracellular matrix deposition, improved myocardial contraction, and calcium homeostasis, suggesting that estradiol directly or indirectly modulates the myocardial transcriptome to preserve cardiovascular function.
Collapse
Affiliation(s)
- Kristofer T. Michalson
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Leanne Groban
- Department of AnesthesiologyWake Forest University School of MedicineWinston‐SalemNC
| | - Timothy D. Howard
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNC
| | - Carol A. Shively
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Areepan Sophonsritsuk
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Susan E. Appt
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - J. Mark Cline
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - Thomas B. Clarkson
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| | - J. Jeffrey Carr
- Department of RadiologyVanderbilt University School of MedicineNashvilleTN
| | - Dalane W. Kitzman
- Section on CardiologyDepartment of Internal MedicineWake Forest University School of MedicineWinston‐SalemNC
| | - Thomas C. Register
- Section on Comparative MedicineDepartment of PathologyWake Forest University School of MedicineWinston‐SalemNC
| |
Collapse
|
35
|
Farré N, Jorba I, Torres M, Falcones B, Martí-Almor J, Farré R, Almendros I, Navajas D. Passive Stiffness of Left Ventricular Myocardial Tissue Is Reduced by Ovariectomy in a Post-menopause Mouse Model. Front Physiol 2018; 9:1545. [PMID: 30455648 PMCID: PMC6230582 DOI: 10.3389/fphys.2018.01545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Heart failure (HF) – a very prevalent disease with high morbidity and mortality – usually presents with diastolic dysfunction. Although post-menopause women are at increased risk of HF and diastolic dysfunction, poor attention has been paid to clinically and experimentally investigate this group of patients. Specifically, whether myocardial stiffness is affected by menopause is unknown. Aim: To investigate whether loss of female sexual hormones modifies the Young’s modulus (E) of left ventricular (LV) myocardial tissue in a mouse model of menopause induced by ovariectomy (OVX). Methods: After 6 months of bilateral OVX, eight mice were sacrificed, fresh LV myocardial strips were prepared (∼8 × 1 × 1 mm), and their passive stress–stretch relationship was measured. E was computed by exponential fitting of the stress–stretch relationship. Subsequently, to assess the relative role of cellular and extracellular matrix components in determining OVX-induced changes in E, the tissues strips were decellularized and subjected to the same stretching protocol to measure E. A control group of eight sham-OVX mice was simultaneously studied. Results: E (kPa; m ± SE) in OVX mice was ∼twofold lower than in controls (11.7 ± 1.8 and 22.1 ± 4.4, respectively; p < 0.05). No significant difference between groups was found in E of the decellularized tissue (31.4 ± 12.05 and 40.9 ± 11.5, respectively; p = 0.58). Conclusion: Loss of female sexual hormones in an OVX model induces a reduction in the passive stiffness of myocardial tissue, suggesting that active relaxation should play a counterbalancing role in diastolic dysfunction in post-menopausal women with HF.
Collapse
Affiliation(s)
- Núria Farré
- Heart Failure Unit, Department of Cardiology, Hospital del Mar, Barcelona, Spain.,Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignasi Jorba
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia - The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marta Torres
- Department of Pneumology, Hospital Clinic Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Bryan Falcones
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Julio Martí-Almor
- Heart Failure Unit, Department of Cardiology, Hospital del Mar, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain
| | - Isaac Almendros
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia - The Barcelona Institute of Science and Technology, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain
| |
Collapse
|
36
|
Wang H, Sun X, Lin MS, Ferrario CM, Van Remmen H, Groban L. G protein-coupled estrogen receptor (GPER) deficiency induces cardiac remodeling through oxidative stress. Transl Res 2018; 199:39-51. [PMID: 29758174 PMCID: PMC6151279 DOI: 10.1016/j.trsl.2018.04.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/09/2018] [Accepted: 04/22/2018] [Indexed: 12/21/2022]
Abstract
Oxidative stress has been implicated in the unfavorable changes in cardiac function and remodeling that occur after ovarian estrogen loss. Using ovariectomized rat models, we previously reported that the cardioprotective actions of estrogen are mediated by the G protein-coupled estrogen receptor (GPER). Here, in 9-month-old, female cardiomyocyte-specific GPER knockout (KO) mice vs sex- and age-matched wild-type (WT) mice, we found increased cardiac oxidative stress and oxidant damage, measured as a decreased ratio of reduced glutathione to oxidized glutathione, increased 4-hydroxynonenal and 8-hydroxy-2'-deoxyguanosine (8-oxo-DG) staining, and increased expression of oxidative stress-related genes. GPER KO mice also displayed increased heart weight, cardiac collagen deposition, and Doppler-derived filling pressure, and decreased percent fractional shortening and early mitral annular velocity compared with WT controls. Treatment of GPER KO mice for 8 weeks with phosphonium [10-(4,5-dimethoxy-2-methyl 3,6-dioxo-1,4-cyclohexadien-1-yl)decyl] triphenyl-,mesylate (MitoQ), a mitochondria-targeted antioxidant, significantly attenuated these measures of cardiac dysfunction, and MitoQ decreased 8-oxo-DG intensity compared with treatment with an inactive comparator compound, (1-decyl)triphenylphosphonium bromide (P <0.05). A real-time polymerase chain reaction array analysis of 84 oxidative stress and antioxidant defense genes revealed that MitoQ attenuates the increase in NADPH oxidase 4 and prostaglandin-endoperoxide synthase 2 and the decrease in uncoupling protein 3 and glutathione S-transferase kappa 1 seen in GPER KO mice. Our findings suggest that the cardioprotective effects of GPER include an antioxidant role and that targeted strategies to limit oxidative stress after early noncancerous surgical extirpation of ovaries or menopause may help limit alterations in cardiac structure and function related to estrogen loss.
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Marina S Lin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma; Biomedical Research Service, Oklahoma City VA Healthcare System, Oklahoma City, Oklahoma
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina; Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina.
| |
Collapse
|
37
|
da Silva JS, Gabriel-Costa D, Wang H, Ahmad S, Sun X, Varagic J, Sudo RT, Ferrario CM, Dell Italia LJ, Sudo GZ, Groban L. Blunting of cardioprotective actions of estrogen in female rodent heart linked to altered expression of cardiac tissue chymase and ACE2. J Renin Angiotensin Aldosterone Syst 2018; 18:1470320317722270. [PMID: 28748720 PMCID: PMC5805468 DOI: 10.1177/1470320317722270] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: Diastolic dysfunction develops in response to hypertension and estrogen (E2) loss and is a forerunner to heart failure (HF) in women. The cardiac renin–angiotensin system (RAS) contributes to diastolic dysfunction, but its role with respect to E2 and blood pressure remain unclear. Methods: We compared the effects of ovariectomy (OVX) or sham surgery on the cardiac RAS, left ventricular (LV) structure/function, and systemic/intracardiac pressures of spontaneously hypertensive rats (SHRs: n = 6 intact and 6 OVX) and age-matched Wistar-Kyoto (WKY: n = 5 intact and 4 OVX) controls. Results: WKY rats were more sensitive to OVX than SHRs with respect to worsening of diastolic function, as reflected by increases in Doppler-derived filling pressures (E/e′) and reductions in myocardial relaxation (e′). This pathobiologic response in WKY rats was directly linked to increases in cardiac gene expression and enzymatic activity of chymase and modest reductions in ACE2 activity. No overt changes in cardiac RAS genes or activities were observed in SHRs, but diastolic function was inversely related to ACE2 activity. Conclusion: Endogenous estrogens exert a more significant regulatory role upon biochemical components of the cardiac RAS of WKY versus SHRs, modulating the lusitropic and structural components of its normotensive phenotype.
Collapse
Affiliation(s)
- Jacqueline S da Silva
- 1 Research Program Development of Drugs, Institute of Biomedical Sciences Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniele Gabriel-Costa
- 1 Research Program Development of Drugs, Institute of Biomedical Sciences Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hao Wang
- 2 The Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA.,3 The Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Sarfaraz Ahmad
- 4 The Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Xuming Sun
- 2 The Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Jasmina Varagic
- 4 The Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Roberto T Sudo
- 1 Research Program Development of Drugs, Institute of Biomedical Sciences Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos M Ferrario
- 4 The Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina, USA.,5 The Department of Internal Medicine-Nephrology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Louis J Dell Italia
- 6 Division of Cardiovascular Disease, University of Alabama at Birmingham and Department of Veterans Affairs, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - Gisele-Zapata Sudo
- 1 Research Program Development of Drugs, Institute of Biomedical Sciences Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leanne Groban
- 2 The Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA.,3 The Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
38
|
Yoshizaki A, Antonio EL, Silva Junior JA, Crajoinas RO, Silva FA, Girardi ACC, Bocalini DS, Portes LA, Dos Santos LFN, Carlos FP, Camillo de Carvalho PDT, Tucci PJF, Serra AJ. Swimming Training Improves Myocardial Mechanics, Prevents Fibrosis, and Alters Expression of Ca2+ Handling Proteins in Older Rats. J Gerontol A Biol Sci Med Sci 2018; 73:468-474. [PMID: 29253100 DOI: 10.1093/gerona/glx244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022] Open
Abstract
Exercise training effects on the contractility of aged myocardium have been investigated for more than 20 years, but the data are still unclear. This study evaluated the hypothesis that a swimming training (ST) may improve myocardial inotropism in older rats. Male Wistar rats aged 4 (young)-and 21 (old)-months-old were divided into young untrained (YNT), old untrained (ONT), and old trained (OTR; 6 weeks of ST) groups. Echocardiography and hemodynamic were employed to assess left ventricular morphology and function. Myocardial mechanics was evaluated on papillary muscles. Histological and immunoblotting were carried out to evaluate fibrosis and proteins that modulate the myocardial function and calcium handling. We found that older rats did not show cardiac dysfunction, but ONT group showed lower physical performance during a swimming test (YNT: 5 ± 2; ONT: -16 ± 0.4; OTR: 51 ± 3; Δ%, sec). Moreover, ONT group showed worse myocardial inotropism, in which it was reversed by ST (Peak developed tension: YNT: 6.2 ± 0.7; ONT: 3.9 ± 0.3; OTR: 6.9 ± 0.9; g/mm2). The ST was associated with preserved collagen content (YNT: 0.38 ± 0.05; ONT: 0.78 ± 0.12; OTR: 0.34 ± 0.09; %). Exercise partially mitigated the effects of aging on intracellular Ca2+-regulating protein (eg, L-Ca2+ channel and phospholamban) and β-isoform of myosin. Thus, we propose that these molecular alterations together with inhibition of collagen increase contribute to improved myocardial performance in older rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Danilo Sales Bocalini
- Center for Physical Education and Sport, Federal University of Espirito Santo, São Paulo, Brazil
| | | | | | | | | | | | - Andrey Jorge Serra
- Federal University of São Paulo, São Paulo, Brazil.,Laboratory of Biophotonic, Nove de Julho University, São Paulo, Brazil
| |
Collapse
|
39
|
Machuki J, Zhang H, Harding S, Sun H. Molecular pathways of oestrogen receptors and β-adrenergic receptors in cardiac cells: Recognition of their similarities, interactions and therapeutic value. Acta Physiol (Oxf) 2018; 222. [PMID: 28994249 PMCID: PMC5813217 DOI: 10.1111/apha.12978] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/07/2017] [Accepted: 10/02/2017] [Indexed: 12/18/2022]
Abstract
Oestrogen receptors (ERs) and β-adrenergic receptors (βARs) play important roles in the cardiovascular system. Moreover, these receptors are expressed in cardiac myocytes and vascular tissues. Numerous experimental observations support the hypothesis that similarities and interactions exist between the signalling pathways of ERs (ERα, ERβ and GPR30) and βARs (β1 AR, β2 AR and β3 AR). The recently discovered oestrogen receptor GPR30 shares structural features with the βARs, and this forms the basis for the interactions and functional overlap. GPR30 possesses protein kinase A (PKA) phosphorylation sites and PDZ binding motifs and interacts with A-kinase anchoring protein 5 (AKAP5), all of which enable its interaction with the βAR pathways. The interactions between ERs and βARs occur downstream of the G-protein-coupled receptor, through the Gαs and Gαi proteins. This review presents an up-to-date description of ERs and βARs and demonstrates functional synergism and interactions among these receptors in cardiac cells. We explore their signalling cascades and the mechanisms that orchestrate their interactions and propose new perspectives on the signalling patterns for the GPR30 based on its structural resemblance to the βARs. In addition, we explore the relevance of these interactions to cell physiology, drugs (especially β-blockers and calcium channel blockers) and cardioprotection. Furthermore, a receptor-independent mechanism for oestrogen and its influence on the expression of βARs and calcium-handling proteins are discussed. Finally, we highlight promising therapeutic avenues that can be derived from the shared pathways, especially the phosphatidylinositol-3-OH kinase (PI3K/Akt) pathway.
Collapse
Affiliation(s)
- J.O. Machuki
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| | - H.Y. Zhang
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| | - S.E. Harding
- National Heart and Lung Institute; Imperial College; London UK
| | - H. Sun
- Department of Physiology; Xuzhou Medical University; Xuzhou China
| |
Collapse
|
40
|
Alencar AK, Montes GC, Montagnoli T, Silva AM, Martinez ST, Fraga AG, Wang H, Groban L, Sudo RT, Zapata-Sudo G. Activation of GPER ameliorates experimental pulmonary hypertension in male rats. Eur J Pharm Sci 2016; 97:208-217. [PMID: 27836751 DOI: 10.1016/j.ejps.2016.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/29/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling that leads to pulmonary congestion, uncompensated right-ventricle (RV) failure, and premature death. Preclinical studies have demonstrated that the G protein-coupled estrogen receptor (GPER) is cardioprotective in male rats and that its activation elicits vascular relaxation in rats of either sex. OBJECTIVES To study the effects of GPER on the cardiopulmonary system by the administration of its selective agonist G1 in male rats with monocrotaline (MCT)-induced PH. METHODS Rats received a single intraperitoneal injection of MCT (60mg/kg) for PH induction. Experimental groups were as follows: control, MCT+vehicle, and MCT+G1 (400μg/kg/daysubcutaneous). Animals (n=5pergroup) were treated with vehicle or G1 for 14days after disease onset. MEASUREMENTS AND MAIN RESULTS Activation of GPER attenuated exercise intolerance and reduced RV overload in PH rats. Rats with PH exhibited echocardiographic alterations, such as reduced pulmonary flow, RV hypertrophy, and left-ventricle dysfunction, by the end of protocol. G1 treatment reversed these PH-related abnormalities of cardiopulmonary function and structure, in part by promoting pulmonary endothelial nitric oxide synthesis, Ca2+ handling regulation and reduction of inflammation in cardiomyocytes, and a decrease of collagen deposition by acting in pulmonary and cardiac fibroblasts. CONCLUSIONS G1 was effective to reverse PH-induced RV dysfunction and exercise intolerance in male rats, a finding that have important implications for ongoing clinical evaluation of new cardioprotective and vasodilator drugs for the treatment of the disease.
Collapse
Affiliation(s)
- Allan K Alencar
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Guilherme C Montes
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tadeu Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ananssa M Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sabrina T Martinez
- Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Aline G Fraga
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hao Wang
- Department of Anesthesiology, Wake Forest University, Winston-Salem, NC, USA
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest University, Winston-Salem, NC, USA
| | - Roberto T Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
41
|
Wang H, Sun X, Chou J, Lin M, Ferrario CM, Zapata-Sudo G, Groban L. Cardiomyocyte-specific deletion of the G protein-coupled estrogen receptor (GPER) leads to left ventricular dysfunction and adverse remodeling: A sex-specific gene profiling analysis. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1870-1882. [PMID: 27725247 DOI: 10.1016/j.bbadis.2016.10.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/13/2016] [Accepted: 10/02/2016] [Indexed: 12/21/2022]
Abstract
Activation of G protein-coupled estrogen receptor (GPER) by its agonist, G1, protects the heart from stressors such as pressure-overload, ischemia, a high-salt diet, estrogen loss, and aging, in various male and female animal models. Due to nonspecific effects of G1, the exact functions of cardiac GPER cannot be concluded from studies using systemic G1 administration. Moreover, global knockdown of GPER affects glucose homeostasis, blood pressure, and many other cardiovascular-related systems, thereby confounding interpretation of its direct cardiac actions. We generated a cardiomyocyte-specific GPER knockout (KO) mouse model to specifically investigate the functions of GPER in cardiomyocytes. Compared to wild type mice, cardiomyocyte-specific GPER KO mice exhibited adverse alterations in cardiac structure and impaired systolic and diastolic function, as measured by echocardiography. Gene deletion effects on left ventricular dimensions were more profound in male KO mice compared to female KO mice. Analysis of DNA microarray data from isolated cardiomyocytes of wild type and KO mice revealed sex-based differences in gene expression profiles affecting multiple transcriptional networks. Gene Set Enrichment Analysis (GSEA) revealed that mitochondrial genes are enriched in GPER KO females, whereas inflammatory response genes are enriched in GPER KO males, compared to their wild type counterparts of the same sex. The cardiomyocyte-specific GPER KO mouse model provides us with a powerful tool to study the functions of GPER in cardiomyocytes. The gene expression profiles of the GPER KO mice provide foundational information for further study of the mechanisms underlying sex-specific cardioprotection by GPER.
Collapse
MESH Headings
- Animals
- Female
- Gene Deletion
- Gene Expression Profiling
- Gene Expression Regulation
- Male
- Mice
- Mice, Knockout
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Sex Characteristics
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Remodeling/genetics
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1009, USA; Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1009, USA
| | - Jeff Chou
- Public Health Sciences, Section on Biostatistical Sciences, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Marina Lin
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1009, USA
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA; Department of Internal Medicine/Nephrology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Gisele Zapata-Sudo
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1009, USA; Institute of Biomedical Sciences, Drug Development Program, Federal University of Rio de Janeiro, Brazil
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1009, USA; Internal Medicine/Molecular Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA; Cardiovascular Research Center, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA; Sticht Center on Aging, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, North Carolina 27157, USA.
| |
Collapse
|