1
|
Li T, Thoen ZE, Applebaum JM, Khalil RA. Menopause-related changes in vascular signaling by sex hormones. J Pharmacol Exp Ther 2025; 392:103526. [PMID: 40184819 DOI: 10.1016/j.jpet.2025.103526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/07/2025] Open
Abstract
Cardiovascular disease (CVD), such as hypertension and coronary artery disease, involves pathological changes in vascular signaling, function, and structure. Vascular signaling is regulated by multiple intrinsic and extrinsic factors that influence endothelial cells, vascular smooth muscle, and extracellular matrix. Vascular function is also influenced by environmental factors including diet, exercise, and stress, as well as genetic background, sex differences, and age. CVD is more common in adult men and postmenopausal women than in premenopausal women. Specifically, women during menopausal transition, with declining ovarian function and production of estrogen (E2) and progesterone, show marked increase in the incidence of CVD and associated vascular dysfunction. Mechanistic research suggests that E2 and E2 receptor signaling have beneficial effects on vascular function including vasodilation, decreased blood pressure, and cardiovascular protection. Also, the tangible benefits of E2 supplementation in improving menopausal symptoms have prompted clinical trials of menopausal hormone therapy (MHT) in CVD, but the results have been inconsistent. The inadequate benefits of MHT in CVD could be attributed to the E2 type, dose, formulation, route, timing, and duration as well as menopausal changes in E2/E2 receptor vascular signaling. Other factors that could affect the responsiveness to MHT are the integrated hormonal milieu including gonadotropins, progesterone, and testosterone, vascular health status, preexisting cardiovascular conditions, and menopause-related dysfunction in the renal, gastrointestinal, endocrine, immune, and nervous systems. Further analysis of these factors should enhance our understanding of menopause-related changes in vascular signaling by sex hormones and provide better guidance for management of CVD in postmenopausal women. SIGNIFICANCE STATEMENT: Cardiovascular disease is more common in adult men and postmenopausal women than premenopausal women. Earlier observations of vascular benefits of menopausal hormone therapy did not materialize in randomized clinical trials. Further examination of the cardiovascular effects of sex hormones in different formulations and regimens, and the menopausal changes in vascular signaling would help to adjust the menopausal hormone therapy protocols in order to enhance their effectiveness in reducing the risk and the management of cardiovascular disease in postmenopausal women.
Collapse
Affiliation(s)
- Tao Li
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Zachary E Thoen
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Jessica M Applebaum
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
2
|
Heydarpour M, Parksook WW, Pojoga LH, Williams GH, Williams JS. Mineralocorticoid Receptor and Aldosterone: Interaction Between NR3C2 Genetic Variants, Sex, and Age in a Mixed Cohort. J Clin Endocrinol Metab 2024; 110:e140-e149. [PMID: 38437868 PMCID: PMC11651684 DOI: 10.1210/clinem/dgae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
CONTEXT Hypertension, a prevalent cardiovascular risk, often involves dysregulated aldosterone and its interaction with the mineralocorticoid receptor (MR). Experimental designs in animal models and human cohorts have demonstrated a sex and age dependency of aldosterone secretion that expands our pathophysiologic understanding. OBJECTIVE This study explores the genetic variation of NR3C2, which encodes MR, in relation to aldosterone, considering age, sex, and race. METHODS Incorporating 720 Caucasians and 145 Africans from the HyperPATH cohort, we investigated the impact of rs4835490, a single nucleotide risk allele variant, on aldosterone levels and vasculature. RESULTS Notably, a significant association between rs4835490 and plasma aldosterone under liberal salt conditions emerged in individuals of European ancestry (P = .0002). Homozygous carriers of the risk A allele exhibited elevated plasma aldosterone levels (AA = 8.1 ± .9 vs GG = 4.9 ± .5 ng/dL). Additionally, aldosterone activation through posture (P = .025) and urinary excretion (P = .0122) showed notable associations. Moreover, genetic interactions with race, sex, and age were observed. Caucasian females under 50 years displayed higher plasma aldosterone, urine aldosterone, and posture aldosterone with the AA genotype compared to females over 50 years, suggesting a potential connection with menopausal or estrogen influences. Interestingly, such age-dependent interactions were absent in the African cohort. CONCLUSION Our study highlights the significance of the NR3C2 genetic variation and its interplay with age, sex, and race in aldosterone activation. The findings point toward an estrogen-modulating effect on MR activation, particularly in women, underlining the role of aldosterone dysregulation in hypertension development. This insight advances our comprehension of hypertension's complexities and opens avenues for personalized interventions. Clinical Trial Registration Number: NCT03029806 (registered January 24, 2017).
Collapse
Affiliation(s)
- Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wasita W Parksook
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine (Division of Endocrinology and Metabolism, and Division of General Internal Medicine), Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Le Nezet E, Marqueze-Pouey C, Guisle I, Clavel MA. Molecular Features of Calcific Aortic Stenosis in Female and Male Patients. CJC Open 2024; 6:1125-1137. [PMID: 39525825 PMCID: PMC11544188 DOI: 10.1016/j.cjco.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/06/2024] [Indexed: 11/16/2024] Open
Abstract
Over the past 15 years, sex-related differences in aortic valve (AV) stenosis (AS) have been highlighted, affecting various aspects of AS, such as the pathophysiology, AV lesions, left ventricle remodelling, and outcomes. Female patients were found to present a more profibrotic pattern of leaflet remodelling and/or thickening, whereas male patients have a preponderance of calcification within stenosed leaflets. The understanding of these sex differences is still limited, owing to the underrepresentation of female patients in many basic and clinical research studies and trials. A better understanding of sex differences in the pathophysiology of AS may highlight new therapeutic targets that potentially could be sex-specific. This review aims to summarize sex-related differences in AS, as discovered from basic research experiments, covering aspects of the disease ranging from leaflet composition to signalling pathways, sex hormones, genetics and/or transcriptomics, and potential sex-adapted medical treatments.
Collapse
Affiliation(s)
- Emma Le Nezet
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Chloé Marqueze-Pouey
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Isabelle Guisle
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et pneumologie de Québec [Quebec Heart & Lung Institute], Université Laval, Québec City, Québec, Canada
| |
Collapse
|
4
|
Carter-Storch R, Le Nezet E, Ali M, Powers A, Haujir A, Demers K, Couture C, Dumont É, Trahan S, Pagé S, Dagenais F, Pibarot P, Dahl JS, Clavel MA. Angiotensin II Receptor Blockers Are Associated With Reduced Valvular Fibrosis in Women With Aortic Stenosis. Can J Cardiol 2024; 40:1690-1699. [PMID: 38518892 DOI: 10.1016/j.cjca.2024.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND Angiotensin receptor blockers (ARBs) may slow down the progression of aortic stenosis (AS) through their antifibrotic effect. Women present more valvular fibrosis than men, so ARBs may have more effect in females. Our aim was to assess the impact of ARBs on the remodelling of the aortic valve in men and women. METHODS We included patients who had an aortic valve replacement with or without coronary bypass grafting from 2006 to 2013. Patients with missing echocardiographic or histologic data were excluded. Warren-Yong and fibrosis scores of the explanted valves were performed. Patients were divided into 4 phenotypes according to their Warren-Yong and fibrosis scores: mild calcification/fibrosis, severe calcification/fibrosis group, predominant fibrosis group, predominant calcification group. RESULTS Among the 1321 included patients, the vast majority (89%) has severe AS. Patients in the predominant fibrosis group, compared with the predominant calcium group, were more often female (39% vs 31%; P = 0.008) with bicuspid valves (44% vs 34%; P = 0.002), and less often used ARBs (25% vs 30%; P = 0.046). Female sex was independently associated with being in the predominant fibrosis group (odds ratio 1.45, 95% confidence interval 1.08-1.95; P = 0.01), with a significant interaction between female sex and ARBs. Women taking ARBs compared with women not taking ARBs had significantly lower fibrosis scores (P < 0.001). This difference was not seen in men. CONCLUSIONS In this large series of patients with moderate-severe AS, among the women there was a negative association between intake of ARBs and valvular fibrosis. Thus, the possible effects of ARBs may be sex specific, with a larger therapeutic role in women.
Collapse
Affiliation(s)
- Rasmus Carter-Storch
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada; Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Emma Le Nezet
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Mulham Ali
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Andréanne Powers
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Amal Haujir
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Karolanne Demers
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Christian Couture
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Éric Dumont
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Sylvain Trahan
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Sylvain Pagé
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - François Dagenais
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada
| | - Jordi S Dahl
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Marie-Annick Clavel
- Institut Universitaire de Cardiologie et de Pneumologie, Université Laval, Québec, Québec, Canada; Department of Cardiology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
5
|
Eissa MA, Gohar EY. Aromatase enzyme: Paving the way for exploring aromatization for cardio-renal protection. Biomed Pharmacother 2023; 168:115832. [PMID: 37931519 PMCID: PMC10843764 DOI: 10.1016/j.biopha.2023.115832] [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: 05/22/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
Documented male-female differences in the risk of cardiovascular and chronic kidney diseases have been largely attributed to estrogens. The cardiovascular and renal protective effects of estrogens are mediated via the activation of estrogen receptors (ERα and ERβ) and G protein-coupled estrogen receptor, and involve interactions with the renin-angiotensin-aldosterone system. Aromatase, also called estrogen synthase, is a cytochrome P-450 enzyme that plays a pivotal role in the conversion of androgens into estrogens. Estrogens are biosynthesized in gonadal and extra-gonadal sites by the action of aromatase. Evidence suggests that aromatase inhibitors, which are used to treat high estrogen-related pathologies, are associated with the development of cardiovascular events. We review the potential role of aromatization in providing cardio-renal protection and highlight several meta-analysis studies on cardiovascular events associated with aromatase inhibitors. Overall, we present the potential of aromatase enzyme as a fundamental contributor to cardio-renal protection.
Collapse
Affiliation(s)
- Manar A Eissa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Merit University, New Sohag, Sohag, Egypt
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
6
|
Stone JC, MacDonald MJ. The impacts of endogenous progesterone and exogenous progestin on vascular endothelial cell, and smooth muscle cell function: A narrative review. Vascul Pharmacol 2023; 152:107209. [PMID: 37591444 DOI: 10.1016/j.vph.2023.107209] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Vascular endothelial and smooth muscle cell dysfunction proceed the development of numerous vascular diseases, such as atherosclerosis. Both estrogen and progesterone receptors are present on vascular endothelial and smooth muscle cells, and therefore it has been postulated that these compounds may affect vascular function. It has been well-established that estrogen is a vasoprotective compound, however, the effects of progesterone on vascular function are not well understood. This narrative review summarizes the current research investigating the impact of both endogenous progesterone, and exogenous synthetic progestin on vascular endothelial and smooth muscle cell function and identifies discrepancies on their effects in vitro and in vivo. We speculate that an inverted-U dose response curve may exist between nitric oxide bioavailability and progesterone concentration, and that the androgenic properties of a progestin may influence vascular function. Future research is needed to discern the effects of both endogenous progesterone and exogenous progestin on vascular endothelial and smooth muscle cell function with consideration for the impacts of progesterone/progestin dose, and progestin type.
Collapse
Affiliation(s)
- Jenna C Stone
- Vascular Dynamics Lab, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Maureen J MacDonald
- Vascular Dynamics Lab, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
7
|
Bernstein SR, Kelleher C, Khalil RA. Gender-based research underscores sex differences in biological processes, clinical disorders and pharmacological interventions. Biochem Pharmacol 2023; 215:115737. [PMID: 37549793 PMCID: PMC10587961 DOI: 10.1016/j.bcp.2023.115737] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Earlier research has presumed that the male and female biology is similar in most organs except the reproductive system, leading to major misconceptions in research interpretations and clinical implications, with serious disorders being overlooked or misdiagnosed. Careful research has now identified sex differences in the cardiovascular, renal, endocrine, gastrointestinal, immune, nervous, and musculoskeletal systems. Also, several cardiovascular, immunological, and neurological disorders have shown differences in prevalence and severity between males and females. Genetic variations in the sex chromosomes have been implicated in several disorders at young age and before puberty. The levels of the gonadal hormones estrogen, progesterone and testosterone and their receptors play a role in the sex differences between adult males and premenopausal women. Hormonal deficiencies and cell senescence have been implicated in differences between postmenopausal and premenopausal women. Specifically, cardiovascular disorders are more common in adult men vs premenopausal women, but the trend is reversed with age with the incidence being greater in postmenopausal women than age-matched men. Gender-specific disorders in females such as polycystic ovary syndrome, hypertension-in-pregnancy and gestational diabetes have attained further research recognition. Other gender-related research areas include menopausal hormone therapy, the "Estrogen Paradox" in pulmonary arterial hypertension being more predominant but less severe in young females, and how testosterone may cause deleterious effects in the kidney while having vasodilator effects in the coronary circulation. This has prompted the National Institutes of Health (NIH) initiative to consider sex as a biological variable in research. The NIH and other funding agencies have provided resources to establish state-of-the-art centers for women health and sex differences in biology and disease in several academic institutions. Scientific societies and journals have taken similar steps to organize specialized conferences and publish special issues on gender-based research. These combined efforts should promote research to enhance our understanding of the sex differences in biological systems beyond just the reproductive system, and provide better guidance and pharmacological tools for the management of various clinical disorders in a gender-specific manner.
Collapse
Affiliation(s)
- Sofia R Bernstein
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Caroline Kelleher
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Yang Y, Li J, Zhou Z, Wu S, Zhao J, Jia W, Liu M, Shen X, He F, Cheng R. Gut Microbiota Perturbation in Early Life Could Influence Pediatric Blood Pressure Regulation in a Sex-Dependent Manner in Juvenile Rats. Nutrients 2023; 15:2661. [PMID: 37375565 DOI: 10.3390/nu15122661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The present study aimed to investigate whether gut dysbiosis induced by ceftriaxone in early life could influence pediatric blood pressure regulation in childhood with or without exposure to a high-fat diet (HFD). Sixty-three newborn pups of Sprague-Dawley rats were administered ceftriaxone sodium or saline solution until weaning at 3 weeks, and the rats were fed a HFD or regular diet from 3 to 6 weeks. Tail-cuff blood pressure, the expression levels of genes of the renin-angiotensin system (RAS), the concentrations of IL-1β, IL-6, and TNF-α in the colon and prefrontal cortex, and the composition of fecal microbiota were analyzed. Ceftriaxone treatment significantly increased the diastolic blood pressure of male rats at 3 weeks. At 6 weeks, systolic blood pressure (SBP) was significantly increased only in ceftriaxone treated male rats fed with HFD. The RAS showed increased activation in the kidney, heart, hypothalamus, and thoracic and abdominal aorta of male rats, but only in the kidney, heart, and hypothalamus of female rats. HFD-fed female rats showed a decreased level of IL-6 in the colon. α diversity of gut microbiota decreased and the Firmicutes to Bacteroidetes ratio increased in both male and female rats at 3 weeks; however, these parameters recovered to various degrees in female rats at 6 weeks. These results revealed that early-life gut dysbiosis induced by antibiotics combined with a HFD in childhood could be involved in pediatric blood pressure regulation and an increase in SBP in juvenile rats, and these effects occurred in a sex-dependent manner.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhimo Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Jincheng Zhao
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Wen Jia
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Meixun Liu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Noto NM, Restrepo YM, Pang HW, Stoyell-Conti F, West CA, Speth RC. Comparative evaluation of biased agonists Sarcosine 1 , d-Alanine 8 -Angiotensin (Ang) II (SD Ang II) and Sarcosine 1 , Isoleucine 8 -Ang II (SI Ang II) and their radioiodinated congeners binding to rat liver membrane AT 1 receptors. Pharmacol Res Perspect 2023; 11:e01053. [PMID: 36639940 PMCID: PMC9840060 DOI: 10.1002/prp2.1053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023] Open
Abstract
Angiotensin II analogue and β-arrestin biased agonist TRV027 (Sarcosine1 , d-Alanine8 -Angiotensin (Ang) II; SD Ang II), developed by Trevena, Inc. in the early 2010s, brought hopes of a novel treatment for cardiovascular diseases, due to its ability to simultaneously cause signaling through the β-arrestin signaling pathway, while antagonizing the pathophysiological effects of Ang II mediated by the AT1 receptor G protein signaling cascades. However, a phase II clinical trial of this agent revealed no significant benefit compared to placebo treatment. Using 125 I-Sarcosine1 , Isoleucine8 -Ang II (125 I-SI Ang II) radioligand receptor competition binding assays, we assessed the relative affinity of TRV027 compared to SI Ang II for liver AT1 receptors. We also compared radioiodinated TRV027 (125 I-SD Ang II) binding affinity for liver AT1 receptors with 125 I-SI Ang II. We found that despite its anticipated gain in metabolic stability, TRV027 and 125 I-SD Ang II had reduced affinity for the AT1 receptor compared with SI Ang II and 125 I-SI Ang II. Additionally, male-female comparisons showed that females have a higher AT1 receptor density, potentially attributed to tissue-dependent estrogen and progesterone effects. Peptide drugs have become more popular over the years due to their increased bioavailability, fast onset of action, high specificity, and low toxicity. Even though Trevena®'s biased agonist peptide TRV027 offered greater stability and potency compared to earlier AT1 R biased agonists, it failed its phase II clinical trial in 2016. Further refinements to AT1 R biased agonist peptides to improve affinity, as seen with SI Ang II, with better stability and bioavailability, has the potential to achieve the anticipated biased agonism.
Collapse
Affiliation(s)
- Natalia M Noto
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Yazmin M Restrepo
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Hong W Pang
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Filipe Stoyell-Conti
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA.,University of Miami, Miami, Florida, USA
| | - Crystal A West
- Department of Biology, Appalachian State University, Kannapolis, North Carolina, USA
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA.,Department of Pharmacology and Physiology, College of Medicine, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
10
|
Iribarren AC, AlBadri A, Wei J, Nelson MD, Li D, Makkar R, Merz CNB. Sex differences in aortic stenosis: Identification of knowledge gaps for sex-specific personalized medicine. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 21:100197. [PMID: 36330169 PMCID: PMC9629620 DOI: 10.1016/j.ahjo.2022.100197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 06/16/2023]
Abstract
Objectives This review summarizes sex-based differences in aortic stenosis (AS) and identifies knowledge gaps that should be addressed by future studies. Background AS is the most common valvular heart disease in developed countries. Sex-specific differences have not been fully appreciated, as a result of widespread under diagnosis of AS in women. Summary Studies including sex-stratified analyses have shown differences in pathophysiology with less calcification and more fibrosis in women's aortic valve. Women have impaired myocardial perfusion reserve and different compensatory response of the left ventricle (LV) to pressure overload, with concentric remodeling and more diffuse fibrosis, in contrast to men with more focal fibrosis and more dilated/eccentrically remodeled LV. There is sex difference in clinical presentation and anatomical characteristics, with women having more paradoxical low-flow/low-gradient AS, under-diagnosis and severity underestimated, with less referral to aortic valve replacement (AVR) compared to men. The response to therapies is also different: women have more adverse events with surgical AVR and greater survival benefit with transcatheter AVR. After AVR, women would have more favorable LV remodeling, but sex-related differences in changes in myocardial reserve flow need future research. Conclusions Investigation into these described sex-related differences in AS offers potential utility for improving prevention and treatment of AS in women and men. To better understand sex-based differences in pathophysiology, clinical presentation, and response to therapies, sex-specific critical knowledge gaps should be addressed in future research for sex-specific personalized medicine.
Collapse
Affiliation(s)
- Ana C. Iribarren
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States of America
| | - Ahmed AlBadri
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States of America
| | - Janet Wei
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States of America
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, CA, United States of America
| | - Michael D. Nelson
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States of America
| | - Debiao Li
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, CA, United States of America
| | - Raj Makkar
- Cedars-Sinai Cardiovascular Intervention Center, Smidt Heart Institute, Los Angeles, CA, United States of America
| | - C. Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States of America
| |
Collapse
|
11
|
Parksook WW, Heydarpour M, Gholami SK, Luther JM, Hopkins PN, Pojoga LH, Williams JS. Salt Sensitivity of Blood Pressure and Aldosterone: Interaction Between the Lysine-specific Demethylase 1 Gene, Sex, and Age. J Clin Endocrinol Metab 2022; 107:1294-1302. [PMID: 35022775 PMCID: PMC9016472 DOI: 10.1210/clinem/dgac011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Indexed: 01/13/2023]
Abstract
CONTEXT Salt sensitivity of blood pressure (SSBP) is associated with increased cardiovascular risk, especially in individuals of African descent, although the underlying mechanisms remain obscure. Lysine-specific demethylase 1 (LSD1) is a salt-sensitive epigenetic regulator associated with SSBP and aldosterone dysfunction. An LSD1 risk allele in humans is associated with SSBP and lower aldosterone levels in hypertensive individuals of African but not European descent. Heterozygous knockout LSD1 mice display SSBP and aldosterone dysregulation, but this effect is modified by age and biological sex. This might explain differences in cardiovascular risk with aging and biological sex in humans. OBJECTIVE This work aims to determine if LSD1 risk allele (rs587618) carriers of African descent display a sex-by-age interaction with SSBP and aldosterone regulation. METHODS We analyzed 297 individuals of African and European descent from the HyperPATH cohort. We performed multiple regression analyses for outcome variables related to SSBP and aldosterone. RESULTS LSD1 risk allele carriers of African (but not European) descent had greater SSBP than nonrisk homozygotes. Female LSD1 risk allele carriers of African descent had greater SSBP, mainly relationship-driven by women with low estrogen (postmenopausal). There was a statistically significant LSD1 genotype-sex interaction in aldosterone response to angiotensin II stimulation in individuals aged 50 years or younger, with female carriers displaying decreased aldosterone responsiveness. CONCLUSION SSBP associated with LSD1 risk allele status is driven by women with a depleted estrogen state. Mechanisms related to a resistance to develop SSBP in females are uncertain but may relate to an estrogen-modulating effect on mineralocorticoid receptor (MR) activation and/or LSD1 epigenetic regulation of the MR.
Collapse
Affiliation(s)
- Wasita W Parksook
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Medicine (Division of Endocrinology and Metabolism, and Division of General Internal Medicine), Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shadi K Gholami
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt Hypertension Center, Nashville, Tennessee 37232, USA
| | - Paul N Hopkins
- Cardiovascular Genetics Research Unit, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
12
|
Lanz-Luces JR, Costa FA, Guzman LFE, Gagliardi ARDT, Lanz-Luces JA, Lanz-Souquett JD, Costa LMAD. DD Genotype and Atherosclerosis in Overweight Menopausal Women. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
13
|
Flores-Monroy J, Lezama-Martínez D, Fonseca-Coronado S, Martínez-Aguilar L. Differences in the expression of the renin angiotensin system and the kallikrein-kinin system during the course of myocardial infarction in male and female Wistar rats. J Renin Angiotensin Aldosterone Syst 2021; 21:1470320319900038. [PMID: 32458737 PMCID: PMC7268575 DOI: 10.1177/1470320319900038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: There is some evidence that components of the renin-angiotensin system and
kallikrein-kinin system are not similarly regulated in both sexes. The aim
of this work was to analyze the expression of angiotensin-converting enzyme,
angiotensin-converting enzyme 2, angiotensin 1 receptor, angiotensin 2
receptor, beta-1 receptor, and beta-2 receptor during the evolution of
myocardial infarction. Methods: Thirty-six male and 36 female Wistar rats were used. Myocardial infarction
was induced. Six groups of both sexes were formed, (n=6):
(a) sham; (b) 48 h myocardial infarction; (c) one week myocardial
infarction; (d) two weeks myocardial infarction; (e) three weeks myocardial
infarction and (f) four weeks myocardial infarction. The expression was
evaluated by real-time polymerase chain reaction on the penumbra of left
ventricle. Results: The mRNA expression of most biomarkers was lower in females than in males.
During acute infarction, an increase of all protein expression was found in
female and at two weeks while in the male only biomarker changes occurred at
three weeks. In addition, in male biomarkers mRNA expression decreased
during chronic infarction while in females it did not. Conclusions: The renin-angiotensin system and kallikrein-kinin system biomarkers
expression occurs at earlier times in the female than in the male rat. In
addition, during chronic myocardial infarction these biomarkers remained
unchanged in females while in males they decreased.
Collapse
Affiliation(s)
- Jazmín Flores-Monroy
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| | - Diego Lezama-Martínez
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| | - Salvador Fonseca-Coronado
- Laboratorio de Inmunobiología de Enfermedades Infecciosas, Universidad Nacional Autonoma de Mexico, Mexico
| | - Luisa Martínez-Aguilar
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| |
Collapse
|
14
|
Czick M, Shapter C, Shapter R. COVID's Razor: RAS Imbalance, the Common Denominator Across Disparate, Unexpected Aspects of COVID-19. Diabetes Metab Syndr Obes 2020; 13:3169-3192. [PMID: 32982349 PMCID: PMC7495349 DOI: 10.2147/dmso.s265518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/04/2020] [Indexed: 12/25/2022] Open
Abstract
A modern iteration of Occam's Razor posits that "the simplest explanation is usually correct." Coronavirus Disease 2019 involves widespread organ damage and uneven mortality demographics, deemed unexpected from what was originally thought to be "a straightforward respiratory virus." The simplest explanation is that both the expected and unexpected aspects of COVID-19 share a common mechanism. Silent hypoxia, atypical acute respiratory distress syndrome (ARDS), stroke, olfactory loss, myocarditis, and increased mortality rates in the elderly, in men, in African-Americans, and in patients with obesity, diabetes, and cancer-all bear the fingerprints of the renin-angiotensin system (RAS) imbalance, suggesting that RAS is the common culprit. This article examines what RAS is and how it works, then from that baseline, the article presents the evidence suggesting RAS involvement in the disparate manifestations of COVID-19. Understanding the deeper workings of RAS helps one make sense of severe COVID-19. In addition, recognizing the role of RAS imbalance suggests potential routes to mitigate COVID-19 severity.
Collapse
Affiliation(s)
- Maureen Czick
- University of Connecticut, Department of Anesthesia, Farmington, CT, USA
| | | | - Robert Shapter
- Independent Consultant ( Medical Research, Medical Communications, and Medical Education), Hartford, CT, USA
| |
Collapse
|
15
|
Sex differences in cardiovascular actions of the renin-angiotensin system. Clin Auton Res 2020; 30:393-408. [PMID: 32860555 DOI: 10.1007/s10286-020-00720-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains a worldwide public health concern despite decades of research and the availability of numerous targeted therapies. While the intrinsic physiological mechanisms regulating cardiovascular function are similar between males and females, marked sex differences have been established in terms of CVD onset, pathophysiology, manifestation, susceptibility, prevalence, treatment responses and outcomes in animal models and clinical populations. Premenopausal females are generally protected from CVD in comparison to men of similar age, with females tending to develop cardiovascular complications later in life following menopause. Emerging evidence suggests this cardioprotection in females is, in part, attributed to sex differences in hormonal regulators, such as the renin-angiotensin system (RAS). To date, research has largely focused on canonical RAS pathways and shown that premenopausal females are protected from cardiovascular derangements produced by activation of angiotensin II pathways. More recently, a vasodilatory arm of the RAS has emerged that is characterized by angiotensin-(1-7) [(Ang-(1-7)], angiotensin-converting enzyme 2 and Mas receptors. Emerging studies provide evidence for a shift towards these cardioprotective Ang-(1-7) pathways in females, with effects modulated by interactions with estrogen. Despite well-established sex differences, female comparison studies on cardiovascular outcomes are lacking at both the preclinical and clinical levels. Furthermore, there are no specific guidelines in place for the treatment of cardiovascular disease in men versus women, including therapies targeting the RAS. This review summarizes current knowledge on sex differences in the cardiovascular actions of the RAS, focusing on interactions with gonadal hormones, emerging data for protective Ang-(1-7) pathways and potential clinical implications for established and novel therapies.
Collapse
|
16
|
Chen J, Lazarenko OP, Zhao H, Wankhade UD, Pedersen K, Watt J, Ronis MJJ. Nox4 Expression Is Not Required for OVX-Induced Osteoblast Senescence and Bone Loss in Mice. JBMR Plus 2020; 4:e10376. [PMID: 32803108 PMCID: PMC7422714 DOI: 10.1002/jbm4.10376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen deficiency and aging play critical roles in the pathophysiology of bone as a result of increased oxidative stress. It has been suggested that prevention of NADPH oxidase- (Nox-) dependent accumulation of ROS may be an approach to potentially minimize bone loss caused by these conditions. Using ovariectomized (OVX) and Nox4 gene-deletion mouse models, we investigated the role of Nox4 in OVX-induced bone loss and osteoblast senescence signaling. Six-month-old WT C57Bl6 mice were allocated to a sham control group, OVX, and OVX plus E2 treatment group for 8 weeks. Decreased bone mass including BMD and BMC were found in the OVX group compared with the sham control (p < 0.05); E2 treatment completely reversed OVX-induced bone loss. Interestingly, the prevention of OVX-induced bone loss by E2 was associated with the elimination of increased senescence signaling in bone osteoblastic cells from the OVX group. E2 blunted OVX-induced p53 and p21 overexpression, but not p16 and Nox4 in bone. In addition, 8- and 11-month-old Nox4 KO female mice were OVX for 8 weeks. Significant bone loss and increased bone osteoblastic cell senescence signaling occurred not only in Nox4 KO OVX mice compared with sham-operated animals, but also in 11-month-old Nox4 KO sham mice compared with 8-month-old Nox4 KO sham mice (p < 0.05). These data suggest that Nox4-mediated ROS in bone osteoblastic cells may be dispensable for sex steroid deficiency-induced bone loss and senescence. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jin‐Ran Chen
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Oxana P Lazarenko
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Haijun Zhao
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Umesh D Wankhade
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Kim Pedersen
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - James Watt
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - Martin J J Ronis
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| |
Collapse
|
17
|
Ramesh SS, Christopher R, Indira Devi B, Bhat DI. The vascular protective role of oestradiol: a focus on postmenopausal oestradiol deficiency and aneurysmal subarachnoid haemorrhage. Biol Rev Camb Philos Soc 2019; 94:1897-1917. [DOI: 10.1111/brv.12541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Shruthi S. Ramesh
- Department of NeurochemistryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Rita Christopher
- Department of NeurochemistryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Bhagavatula Indira Devi
- Department of NeurosurgeryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| | - Dhananjaya I. Bhat
- Department of NeurosurgeryNational Institute of Mental Health and Neuro Sciences Bengaluru‐560029 Karnataka India
| |
Collapse
|
18
|
Stanhewicz AE, Wenner MM, Stachenfeld NS. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. Am J Physiol Heart Circ Physiol 2018; 315:H1569-H1588. [PMID: 30216121 PMCID: PMC6734083 DOI: 10.1152/ajpheart.00396.2018] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/22/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
Diseases of the cardiovascular system are the leading cause of morbidity and mortality in men and women in developed countries, and cardiovascular disease (CVD) is becoming more prevalent in developing countries. The prevalence of atherosclerotic CVD in men is greater than in women until menopause, when the prevalence of CVD increases in women until it exceeds that of men. Endothelial function is a barometer of vascular health and a predictor of atherosclerosis that may provide insights into sex differences in CVD as well as how and why the CVD risk drastically changes with menopause. Studies of sex differences in endothelial function are conflicting, with some studies showing earlier decrements in endothelial function in men compared with women, whereas others show similar age-related declines between the sexes. Because the increase in CVD risk coincides with menopause, it is generally thought that female hormones, estrogens in particular, are cardioprotective. Moreover, it is often proposed that androgens are detrimental. In truth, the relationships are more complex. This review first addresses female and male sex hormones and their receptors and how these interact with the cardiovascular system, particularly the endothelium, in healthy young women and men. Second, we address sex differences in sex steroid receptor-independent mechanisms controlling endothelial function, focusing on vascular endothelin and the renin-angiotensin systems, in healthy young women and men. Finally, we discuss sex differences in age-associated endothelial dysfunction, focusing on the role of attenuated circulating sex hormones in these effects.
Collapse
Affiliation(s)
- Anna E Stanhewicz
- Department of Kinesiology, Pennsylvania State University , University Park, Pennsylvania
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| | - Nina S Stachenfeld
- The John B. Pierce Laboratory, New Haven, Connecticut
- Department of Obstetrics, Gynecology and Reproductive Sciences and Yale School of Public Health, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
19
|
de Almeida SA, Claudio ERG, Mengal V, Brasil GA, Merlo E, Podratz PL, Graceli JB, Gouvea SA, de Abreu GR. Estrogen Therapy Worsens Cardiac Function and Remodeling and Reverses the Effects of Exercise Training After Myocardial Infarction in Ovariectomized Female Rats. Front Physiol 2018; 9:1242. [PMID: 30233413 PMCID: PMC6134041 DOI: 10.3389/fphys.2018.01242] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/16/2018] [Indexed: 01/24/2023] Open
Abstract
There is an increase in the incidence of cardiovascular events such as myocardial infarction (MI) after menopause. However, the use of estrogen therapy (E2) remains controversial. The aim of this study was to evaluate the effects of E2, alone and combined with exercise training (ET), on cardiac function and remodeling in ovariectomized (OVX) rats after MI. Wistar female rats underwent ovariectomy, followed by MI induction were separated into five groups: S; MI; MI+ET; MI+E2; and MI+ET+E2. Fifteen days after MI or sham surgery, treadmill ET and/or estrogen therapy [17-β estradiol-3-benzoate (E2), s.c. three times/week] were initiated and maintained for 8 weeks. After the treatment and/or training period, the animals underwent cardiac hemodynamic evaluation through catheterization of the left ventricle (LV); the LV systolic and diastolic pressures (LVSP and LVEDP, respectively), maximum LV contraction and relaxation derivatives (dP/dt+ and dP/dt−), and isovolumic relaxation time (Tau) were assessed. Moreover, histological analyses of the heart (collagen and hypertrophy), cardiac oxidative stress [advanced oxidation protein products (AOPPs)], pro- and antioxidant protein expression by Western blotting and antioxidant enzyme activity in the heart were evaluated. The MI reduced the LVSP, dP/dt+ and dP/dt− but increased the LVEDP and Tau. E2 did not prevent the MI-induced changes in cardiac function, even when combined with ET. An increase in the dP/dt+ was observed in the E2 group compared with the MI group. There were no changes in collagen deposition and myocyte hypertrophy caused by the treatments. The increases in AOPP, gp91-phox, and angiotensin II type 1 receptor expression induced by MI were not reduced by E2. There were no changes in the expression of catalase caused by MI or by the treatments, although, a reduction in superoxide dismutase (SOD) expression occurred in the groups subjected to E2 treatment. Whereas there were post-MI reductions in activities of SOD and catalase enzymes, only that of SOD was prevented by ET. Therefore, we conclude that E2 therapy does not prevent the MI-induced changes in cardiac function and worsens parameters related to cardiac remodeling. Moreover, E2 reverses the positive effects of ET when used in combination, in OVX infarcted female rats.
Collapse
Affiliation(s)
- Simone Alves de Almeida
- Laboratório de Regulação Neurohumoral da Circulação, Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Erick R G Claudio
- Laboratório de Regulação Neurohumoral da Circulação, Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Vinicius Mengal
- Laboratório de Regulação Neurohumoral da Circulação, Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Girlandia A Brasil
- Núcleo de Pesquisas em Ciências Farmacêuticas - Nupecfarma, Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Vila Velha, Vila Velha, Brazil
| | - Eduardo Merlo
- Departamento de Morfologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Priscila L Podratz
- Departamento de Morfologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Jones B Graceli
- Departamento de Morfologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Sonia A Gouvea
- Laboratório de Regulação Neurohumoral da Circulação, Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Gláucia Rodrigues de Abreu
- Laboratório de Regulação Neurohumoral da Circulação, Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, Brazil
| |
Collapse
|
20
|
Al-Shboul OA, Al-Dwairi AN, Alqudah MA, Mustafa AG. Gender differences in the regulation of MLC 20 phosphorylation and smooth muscle contraction in rat stomach. Biomed Rep 2018; 8:283-288. [PMID: 29599980 DOI: 10.3892/br.2018.1053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/19/2018] [Indexed: 01/03/2023] Open
Abstract
Evidence of sex-related differences in gastrointestinal (GI) functions has been reported in the literature. In addition, various GI disorders have disproportionate prevalence between the sexes. An essential step in the initiation of smooth muscle contraction is the phosphorylation of the 20-kDa regulatory myosin light chain (MLC20) by the Ca2+/calmodulin-dependent myosin light chain kinase (MLCK). However, whether male stomach smooth muscle inherits different contractile signaling mechanisms for the regulation of MLC20 phosphorylation from that in females has not been established. The present study was designed to investigate sex-associated differences in the regulation of MLC20 phosphorylation and thus muscle contraction in gastric smooth muscle cells (GSMCs). Experiments were performed on GSMCs freshly isolated from male and female rats. Contraction of the GSMCs in response to acetylcholine (ACh), a muscarinic agonist, was measured via scanning micrometry in the presence or absence of the MLCK inhibitor, ML-7. Additionally, the protein levels of MLC20, MLCK and phosphorylated MLC20 were measured by ELISA. The protein levels of MLC20 and MLCK were indifferent between the sexes. ACh induced greater contraction (P<0.05) as well as greater MLC20 phosphorylation (P<0.05) in male GSMCs compared with female. Pretreatment of GSMCs with ML-7 significantly reduced the ACh-induced contraction (P<0.05) and MLC20 phosphorylation (P<0.05) in the male and female cells, and notably, abolished the contractile differences between the sexes. In conclusion, MLC20 phosphorylation and thus muscle contraction may be activated to a greater extent in male rat stomach compared with that in females.
Collapse
Affiliation(s)
- Othman A Al-Shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmed N Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad A Alqudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ayman G Mustafa
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
21
|
Colafella KMM, Denton KM. Sex-specific differences in hypertension and associated cardiovascular disease. Nat Rev Nephrol 2018; 14:185-201. [PMID: 29380817 DOI: 10.1038/nrneph.2017.189] [Citation(s) in RCA: 316] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although intrinsic mechanisms that regulate arterial blood pressure (BP) are similar in men and women, marked variations exist at the molecular, cellular and tissue levels. These physiological disparities between the sexes likely contribute to differences in disease onset, susceptibility, prevalence and treatment responses. Key systems that are important in the development of hypertension and cardiovascular disease (CVD), including the sympathetic nervous system, the renin-angiotensin-aldosterone system and the immune system, are differentially activated in males and females. Biological age also contributes to sexual dimorphism, as premenopausal women experience a higher degree of cardioprotection than men of similar age. Furthermore, sex hormones such as oestrogen and testosterone as well as sex chromosome complement likely contribute to sex differences in BP and CVD. At the cellular level, differences in cell senescence pathways may contribute to increased longevity in women and may also limit organ damage caused by hypertension. In addition, many lifestyle and environmental factors - such as smoking, alcohol consumption and diet - may influence BP and CVD in a sex-specific manner. Evidence suggests that cardioprotection in women is lost under conditions of obesity and type 2 diabetes mellitus. Treatment strategies for hypertension and CVD that are tailored according to sex could lead to improved outcomes for affected patients.
Collapse
Affiliation(s)
- Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University Wellington Road, Clayton, Victoria 3800, Australia.,Department of Physiology, Monash University, 26 Innovation Walk, Clayton, Victoria 3800, Australia.,Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, Netherlands
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University Wellington Road, Clayton, Victoria 3800, Australia.,Department of Physiology, Monash University, 26 Innovation Walk, Clayton, Victoria 3800, Australia
| |
Collapse
|
22
|
Zhao Y, Zhu Q, Sun S, Qiu Y, Li J, Liu W, Yuan G, Ma H. Renal transplantation increases angiotensin II receptor-mediated vascular contractility associated with changes of epigenetic mechanisms. Int J Mol Med 2018; 41:2375-2388. [PMID: 29393347 DOI: 10.3892/ijmm.2018.3435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2017] [Indexed: 11/06/2022] Open
Abstract
Hypertension is one of the most common complications following renal transplantation, and it increases the risk of graft loss and other cardiovascular diseases. Previous studies have revealed that the use of angiotensin II (Ang II) blockers for preventing and treating hypertension is closely associated with higher survival following renal transplantation. However, the cellular and molecular mechanisms by which the vascular contractility of the recipient is altered in response to Ang II following renal transplantation have not been fully elucidated. In the present study, using the Fisher‑Lewis rat kidney transplantation model, the blood pressure (BP) of the conscious transplant recipient was measured following the intravenous administration of Ang II. In addition, the mechanisms underlying the Ang II-mediated vascular contractility via the type 1 and type 2 Ang II receptors (AT1R and AT2R, respectively) in large and small-resistance blood vessels were determined in the recipient after renal transplantation. The results showed that renal transplantation significantly increased the Ang II-stimulated BP of the rats. Additionally, ex vivo contractility experiments using aorta and mesenteric arteries revealed that the contractions induced by Ang II were significantly strengthened in the recipient following renal transplantation, and were associated with an increased intracellular Ca2+ concentration. Losartan almost eradicated the Ang II-induced contractions whereas PD-123319 had no apparent effects on the Ang II-induced contractions in the aorta and mesenteric arteries of the recipient. Furthermore, the expression levels of AT1R but not AT2R were significantly increased in the vasculature of the recipient following renal transplantation, which exhibited a close association with selective DNA demethylation detected in the promoter region of the vascular AT1aR gene. These results indicate that changes of recipient vascular AT1R gene expression, occurring through a mechanism involving DNA methylation, increase the vascular contractility in response to Ang II. This may lead to the increased risk of hypertension following renal transplantation.
Collapse
Affiliation(s)
- Yakun Zhao
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Qingguo Zhu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shiping Sun
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Qiu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jingquan Li
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wei Liu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Gangjun Yuan
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hua Ma
- Surgical Department, The People's Hospital of Fuyun County, Aletai, Xinjiang 836100, P.R. China
| |
Collapse
|
23
|
Asadian S, Siavashi V, Jabarpour M, Sharifi A, Esmaeilivand M, Pourmohammad P, Nassiri SM. Circulating endothelial progenitor cells in pregnant women with premature rupture of membranes: potential association with placental disorders. Reprod Fertil Dev 2018; 30:1689-1698. [DOI: 10.1071/rd17523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 05/09/2018] [Indexed: 02/03/2023] Open
Abstract
The frequency of preterm labour has risen over the last few years. Plasma oestrogen concentrations differ between patients who deliver before term and those who deliver at term. Oestrogen can influence the kinetics of circulating endothelial progenitor cells (cEPCs). Here, we attempted to identify the potential association of cEPCs with the incidence of complications typical of prematurity. The study groups consisted of 60 pregnant women with premature rupture of membranes (PROM; less than 37 weeks) and 50 term pregnant women (more than 38 weeks). cEPCs were isolated from term pregnant women and pregnant women with PROM and then migratory, proliferative, tubulogenic and functional properties of these cells along with serum secretion of important EPC chemotactic cytokines were analysed. In addition, the effect of 17β-oestradiol on biological features of cEPCs harvested from pregnant women was investigated. Our results showed that an increased concentration of oestrogen in women with PROM was associated with increased numbers of cEPCs, with these cells having increased oestrogen receptor α expression together with augmented proliferative, migratory and colony-formation properties. 17β-oestradiol induced proliferation, migration and angiogenic secretory activity of cEPCs from pregnant women. Overall, circulation mobilisation of EPCs in pregnant women may be associated with placental disorders.
Collapse
|
24
|
Nehme A, Zibara K. Cellular distribution and interaction between extended renin-angiotensin-aldosterone system pathways in atheroma. Atherosclerosis 2017; 263:334-342. [PMID: 28600074 DOI: 10.1016/j.atherosclerosis.2017.05.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/14/2017] [Accepted: 05/24/2017] [Indexed: 01/06/2023]
Abstract
The importance of the renin-angiotensin-aldosterone system (RAAS) in the development of atherosclerotic has been experimentally documented. In fact, RAAS components have been shown to be locally expressed in the arterial wall and to be differentially regulated during atherosclerotic lesion progression. RAAS transcripts and proteins were shown to be differentially expressed and to interact in the 3 main cells of atheroma: endothelial cells, vascular smooth muscle cells, and macrophages. This review describes the local expression and cellular distribution of extended RAAS components in the arterial wall and their differential regulation during atherosclerotic lesion development.
Collapse
Affiliation(s)
- Ali Nehme
- EA4173, Functional Genomics of Arterial Hypertension, Hôpital Nord-Ouest, Villefranche-sur-Saône, Université Lyon1, Lyon, France; ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
25
|
Spaanderman MEA, Ekhart THA, de Leeuw PW, Peeters LLH. Angiotensin II Sensitivity in Nonpregnant Formerly Preeclamptic Women and Halthy Parous Contorls. ACTA ACUST UNITED AC 2016; 11:416-22. [PMID: 15350256 DOI: 10.1016/j.jsgi.2004.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND In women prone to develop hypertensive complications, vascular reactivity fails to decrease in early pregnancy. Since hypertensive syndromes of pregnancy seem to be superimposed on a preexisting disorder, we tested the hypothesis that in formerly preeclamptic women, as compared to healthy parous controls, circulatory reactivity to angiotensin II is enhanced in the follicular phase of the menstrual cycle. METHODS Sixty formerly preeclamptic women were subdivided into a hypertensive (HYPERT, n = 14), a normotensive thrombophilic (THROMB, n = 26), and a normotensive nonthrombophilic (ASYMPT, n = 20) subgroup. In these women and in 11 healthy parous controls we assessed at least 5 months postpartum at day 5 (+/-2) of the menstrual cycle the following variables: body weight, height, plasma volume, reactivity to infused angiotensin II of arterial blood pressure, heart rate, glomerular filtration rate (GFR), effective renal blood flow, and the hormones of the renin-angiotensin-aldosterone (RAAS) axis. RESULTS At baseline, THROMB did not differ from controls. In contrast, ASYMPT exhibited slight overweight, reduced plasma volume, and reduced renal blood flow. HYPERT much resembled ASYMPT except for the overweight, renal perfusion, and GFR. Infusion of angiotensin II led to comparable decreases in renal perfusion and filtration, and to increases in blood pressure. However, sensitivity to this substance correlated inversely with relative and absolute sizes of the plasma volume compartment. CONCLUSION Circulatory sensitivity to infused angiotensin II is comparable between nonpregnant formerly preeclamptic women and healthy parous controls. However, responsiveness to this agent is enhanced among women with a contracted plasma volume compartment, a condition commonly observed among formerly preeclamptic women.
Collapse
|
26
|
Yang X, Zhang W, Chen Y, Li Y, Sun L, Liu Y, Liu M, Yu M, Li X, Han J, Duan Y. Activation of Peroxisome Proliferator-activated Receptor γ (PPARγ) and CD36 Protein Expression: THE DUAL PATHOPHYSIOLOGICAL ROLES OF PROGESTERONE. J Biol Chem 2016; 291:15108-18. [PMID: 27226602 DOI: 10.1074/jbc.m116.726737] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Indexed: 12/27/2022] Open
Abstract
Progesterone or its analog, one of components of hormone replacement therapy, may attenuate the cardioprotective effects of estrogen. However, the underlying mechanisms have not been fully elucidated. Expression of CD36, a receptor for oxidized LDL (oxLDL) that enhances macrophage/foam cell formation, is activated by the transcription factor peroxisome proliferator-activated receptor γ (PPARγ). CD36 also functions as a fatty acid transporter to influence fatty acid metabolism and the pathophysiological status of several diseases. In this study, we determined that progesterone induced macrophage CD36 expression, which is related to progesterone receptor (PR) activity. Progesterone enhanced cellular oxLDL uptake in a CD36-dependent manner. Mechanistically, progesterone increased PPARγ expression and PPARγ promoter activity in a PR-dependent manner and the binding of PR with the progesterone response element in the PPARγ promoter. Specific deletion of macrophage PPARγ (MφPPARγ KO) expression in mice abolished progesterone-induced macrophage CD36 expression and cellular oxLDL accumulation. We also determined that, associated with gestation and increased serum progesterone levels, CD36 and PPARγ expression in mouse adipose tissue, skeletal muscle, and peritoneal macrophages were substantially activated. Taken together, our study demonstrates that progesterone can play dual pathophysiological roles by activating PPARγ expression, in which progesterone increases macrophage CD36 expression and oxLDL accumulation, a negative effect on atherosclerosis, and enhances the PPARγ-CD36 pathway in adipose tissue and skeletal muscle, a protective effect on pregnancy.
Collapse
Affiliation(s)
| | | | - Yuanli Chen
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China School of Medicine, and
| | - Yan Li
- From the College of Life Sciences
| | - Lei Sun
- From the College of Life Sciences
| | - Ying Liu
- From the College of Life Sciences
| | | | - Miao Yu
- From the College of Life Sciences
| | | | - Jihong Han
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| | - Yajun Duan
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| |
Collapse
|
27
|
Menazza S, Murphy E. The Expanding Complexity of Estrogen Receptor Signaling in the Cardiovascular System. Circ Res 2016; 118:994-1007. [PMID: 26838792 DOI: 10.1161/circresaha.115.305376] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Estrogen has important effects on cardiovascular function including regulation of vascular function, blood pressure, endothelial relaxation, and the development of hypertrophy and cardioprotection. However, the mechanisms by which estrogen mediates these effects are still poorly understood. As detailed in this review, estrogen can regulate transcription by binding to 2 nuclear receptors, ERα and ERβ, which differentially regulate gene transcription. ERα and ERβ regulation of gene transcription is further modulated by tissue-specific coactivators and corepressors. Estrogen can bind to ERα and ERβ localized at the plasma membrane as well as G-protein-coupled estrogen receptor to initiate membrane delimited signaling, which enhances kinase signaling pathways that can have acute and long-term effects. The kinase signaling pathways can also mediate transcriptional changes and can synergize with the ER to regulate cell function. This review will summarize the beneficial effects of estrogen in protecting the cardiovascular system through ER-dependent mechanisms with an emphasis on the role of the recently described ER membrane signaling mechanisms.
Collapse
Affiliation(s)
- Sara Menazza
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Elizabeth Murphy
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
28
|
|
29
|
Sathishkumar K, Balakrishnan MP, Yallampalli C. Enhanced mesenteric arterial responsiveness to angiotensin II is androgen receptor-dependent in prenatally protein-restricted adult female rat offspring. Biol Reprod 2014; 92:55. [PMID: 25550341 DOI: 10.1095/biolreprod.114.126482] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Gestational protein restriction results in intrauterine growth restriction and hypertension in adult female growth-restricted rats. Enhanced vascular responsiveness to angiotensin II is observed, and blockade of the renin-angiotensin system abolishes hypertension in adult growth-restricted rats, suggesting that the renin-angiotensin system contributes to intrauterine growth restriction-induced hypertension. Moreover, growth-restricted adult rats have higher plasma testosterone levels, and antiandrogen treatment abolishes hypertension, indicating an important role for testosterone. We hypothesized that androgens may play a pivotal role in the enhanced responsiveness to Ang II and hypertension. Female offspring of pregnant rats fed 20% protein (control) or 6% protein diet (protein restricted), at 6 mo of age, were studied. Plasma testosterone and mean arterial pressure in protein-restricted offspring were significantly higher compared to controls. Flutamide treatment (10 mg/kg/day subcutaneously for 10 days) reduced mean arterial pressure in protein-restricted offspring but was without significant effect in controls. Vascular Agtr1/Agtr2 ratio was significantly higher in protein-restricted offspring, an effect that was reversed by flutamide. Flutamide treatment did not have any effect on Agtr1/Agtr2 ratio in controls. Enhanced contractile response to angiotensin II in mesenteric arteries was observed in protein-restricted offspring compared with control. Flutamide treatment reversed the enhanced contractile response to angiotensin II in protein-restricted offspring without significant effect in controls. Vascular reactivity to phenylephrine was similar between the control and protein-restricted offspring with and without flutamide treatment, suggesting that enhanced contractile response and flutamide's reversal effect is specific to angiotensin II. These results suggest that prenatally protein-restricted rats exhibit an enhanced responsiveness to angiotensin II that is testosterone-dependent.
Collapse
Affiliation(s)
- Kunju Sathishkumar
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, Texas
| | - Meena P Balakrishnan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
30
|
Xiao D, Dasgupta C, Li Y, Huang X, Zhang L. Perinatal nicotine exposure increases angiotensin II receptor-mediated vascular contractility in adult offspring. PLoS One 2014; 9:e108161. [PMID: 25265052 PMCID: PMC4179262 DOI: 10.1371/journal.pone.0108161] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 08/18/2014] [Indexed: 12/18/2022] Open
Abstract
Previous studies have reported that perinatal nicotine exposure causes development of hypertensive phenotype in adult offspring. Aims The present study was to determine whether perinatal nicotine exposure causes an epigenetic programming of vascular Angiotensin II receptors (ATRs) and their-mediated signaling pathway leading to heightened vascular contraction in adult offspring. Main methods Nicotine was administered to pregnant rats via subcutaneous osmotic minipumps from day 4 of gestation to day 10 after birth. The experiments were conducted at 5 months of age of male offspring. Key Findings Nicotine treatment enhanced Angitension II (Ang II)-induced vasoconstriction and 20-kDa myosin light chain phosphorylation (MLC20-P) levels. In addition, the ratio of Ang II-induced tension/MLC-P was also significantly increased in nicotine-treated group compared with the saline group. Nicotine-mediated enhanced constrictions were not directly dependent on the changes of [Ca2+]i concentrations but dependent on Ca2+ sensitivity. Perinatal nicotine treatment significantly enhanced vascular ATR type 1a (AT1aR) but not AT1bR mRNA levels in adult rat offspring, which was associated with selective decreases in DNA methylation at AT1aR promoter. Contrast to the effect on AT1aR, nicotine decreased the mRNA levels of vascular AT2R gene, which was associated with selective increases in DNA methylation at AT2R promoter. Significance Our results indicated that perinatal nicotine exposure caused an epigenetic programming of vascular ATRs and their-mediated signaling pathways, and suggested that differential regulation of AT1R/AT2R gene expression through DNA methylation mechanism may be involved in nicotine-induced heightened vasoconstriction and development of hypertensive phenotype in adulthood.
Collapse
Affiliation(s)
- DaLiao Xiao
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
- * E-mail:
| | - Chiranjib Dasgupta
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Yong Li
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Xiaohui Huang
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Lubo Zhang
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| |
Collapse
|
31
|
Chinnathambi V, More AS, Hankins GD, Yallampalli C, Sathishkumar K. Gestational exposure to elevated testosterone levels induces hypertension via heightened vascular angiotensin II type 1 receptor signaling in rats. Biol Reprod 2014; 91:6. [PMID: 24855104 DOI: 10.1095/biolreprod.114.118968] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pre-eclampsia is a life-threatening pregnancy disorder whose pathogenesis remains unclear. Plasma testosterone levels are elevated in pregnant women with pre-eclampsia and polycystic ovary syndrome, who often develop gestational hypertension. We tested the hypothesis that increased gestational testosterone levels induce hypertension via heightened angiotensin II signaling. Pregnant Sprague-Dawley rats were injected with vehicle or testosterone propionate from Gestational Day 15 to 19 to induce a 2-fold increase in plasma testosterone levels, similar to levels observed in clinical conditions like pre-eclampsia. A subset of rats in these two groups was given losartan, an angiotensin II type 1 receptor antagonist by gavage during the course of testosterone exposure. Blood pressure levels were assessed through a carotid arterial catheter and endothelium-independent vascular reactivity through wire myography. Angiotensin II levels in plasma and angiotensin II type 1 receptor expression in mesenteric arteries were also examined. Blood pressure levels were significantly higher on Gestational Day 20 in testosterone-treated dams than in controls. Treatment with losartan during the course of testosterone exposure significantly attenuated testosterone-induced hypertension. Plasma angiotensin II levels were not significantly different between control and testosterone-treated rats; however, elevated testosterone levels significantly increased angiotensin II type 1 receptor protein levels in the mesenteric arteries. In testosterone-treated rats, mesenteric artery contractile responses to angiotensin II were significantly greater, whereas contractile responses to K(+) depolarization and phenylephrine were unaffected. The results demonstrate that elevated testosterone during gestation induces hypertension in pregnant rats via heightened angiotensin II type 1 receptor-mediated signaling, providing a molecular mechanism linking elevated maternal testosterone levels with gestational hypertension.
Collapse
Affiliation(s)
- Vijayakumar Chinnathambi
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Amar S More
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Gary D Hankins
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas
| | - Chandra Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas
| | - Kunju Sathishkumar
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
32
|
Koledova VV, Khalil RA. Sex hormone replacement therapy and modulation of vascular function in cardiovascular disease. Expert Rev Cardiovasc Ther 2014; 5:777-89. [PMID: 17605655 DOI: 10.1586/14779072.5.4.777] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epidemiological and experimental studies suggest vascular protective effects of estrogen. Cardiovascular disease (CVD) is less common in premenopausal women than in men and postmenopausal women. Cytosolic/nuclear estrogen receptors (ERs) have been shown to mediate genomic effects that stimulate endothelial cell growth but inhibit vascular smooth muscle proliferation. However, the Heart and Estrogen/Progestin Replacement Study (HERS), HERS-II and Women's Health Initiative clinical trials demonstrated that hormone replacement therapy (HRT) may not provide vascular benefits in postmenopausal women and may instead trigger adverse cardiovascular events. HRT may not provide vascular benefits because of the type of hormone used. Oral estrogens are biologically transformed by first-pass metabolism in the liver. By contrast, transdermal preparations avoid first pass metabolism. Also, natural estrogens and phytoestrogens may provide alternatives to synthetic estrogens. Furthermore, specific ER modulators could minimize the adverse effects of HRT, including breast cancer. HRT failure in CVD could also be related to changes in vascular ERs. Genetic polymorphism and postmenopausal decrease in vascular ERs or the downstream signaling mechanisms may reduce the effects of HRT. HRT in the late postmenopausal period may not be as effective as during menopausal transition. Additionally, while HRT may aggravate pre-existing CVD, it may thwart its development if used in a timely fashion. Lastly, the vascular effects of progesterone and testosterone, as well as modulators of their receptors, may modify the effects of estrogen and thereby provide alternative HRT strategies. Thus, the beneficial effects of HRT in postmenopausal CVD can be enhanced by customizing the HRT type, dose, route of administration and timing depending on the subject's age and cardiovascular condition.
Collapse
Affiliation(s)
- Vera V Koledova
- Brigham and Women's Hospital, Division of Vascular Surgery, 75 Francis Street, Boston, MA 02115, USA.
| | | |
Collapse
|
33
|
Khalil RA. Estrogen, vascular estrogen receptor and hormone therapy in postmenopausal vascular disease. Biochem Pharmacol 2013; 86:1627-42. [PMID: 24099797 DOI: 10.1016/j.bcp.2013.09.024] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is less common in premenopausal women than men of the same age or postmenopausal women, suggesting vascular benefits of estrogen. Estrogen activates estrogen receptors ERα, ERβ and GPR30 in endothelium and vascular smooth muscle (VSM), which trigger downstream signaling pathways and lead to genomic and non-genomic vascular effects such as vasodilation, decreased VSM contraction and growth and reduced vascular remodeling. However, randomized clinical trials (RCTs), such as the Women's Health Initiative (WHI) and Heart and Estrogen/progestin Replacement Study (HERS), have shown little vascular benefits and even adverse events with menopausal hormone therapy (MHT), likely due to factors related to the MHT used, ER profile, and RCT design. Some MHT forms, dose, combinations or route of administration may have inadequate vascular effects. Age-related changes in ER amount, distribution, integrity and post-ER signaling could alter the vascular response to MHT. The subject's age, preexisting CVD, and hormone environment could also reduce the effects of MHT. Further evaluation of natural and synthetic estrogens, phytoestrogens, and selective estrogen-receptor modulators (SERMs), and the design of appropriate MHT combinations, dose, route and 'timing' could improve the effectiveness of conventional MHT and provide alternative therapies in the peri-menopausal period. Targeting ER using specific ER agonists, localized MHT delivery, and activation of specific post-ER signaling pathways could counter age-related changes in ER. Examination of the hormone environment and conditions associated with hormone imbalance such as polycystic ovary syndrome may reveal the causes of abnormal hormone-receptor interactions. Consideration of these factors in new RCTs such as the Kronos Early Estrogen Prevention Study (KEEPS) could enhance the vascular benefits of estrogen in postmenopausal CVD.
Collapse
Affiliation(s)
- Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Thomas P, Pang Y. Protective actions of progesterone in the cardiovascular system: potential role of membrane progesterone receptors (mPRs) in mediating rapid effects. Steroids 2013; 78:583-8. [PMID: 23357432 DOI: 10.1016/j.steroids.2013.01.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/04/2013] [Accepted: 01/14/2013] [Indexed: 02/06/2023]
Abstract
The protective functions of progesterone in the cardiovascular system have received little attention even though evidence has accumulated that progesterone lowers blood pressure, inhibits coronary hyperactivity and has powerful vasodilatory and natriuretic effects. One possible reason why potential beneficial actions of progesterone on cardiovascular functions have not been extensively studied is that divergent effects to those of progesterone have been observed in many clinical trials with synthetic progestins such as medroxyprogesterone acetate which are associated with increased risk of coronary disease. Evidence that progesterone exerts protective effects on cardiovascular functions is briefly reviewed. The finding that progesterone administration decreases blood vessel vasoconstriction in several animal models within a few minutes suggests that rapid, nongenomic progesterone mechanisms are of physiological importance in regulating vascular tone. Rapid activation of second messenger pathways by progesterone has been observed in vascular endothelial and smooth muscle cells, resulting in alterations in endothelial nitric oxide synthase (eNOS) activity and calcium influx, respectively. Both nuclear progesterone receptors (PRs) and novel membrane progesterone receptors (mPRs) are candidates for the intermediaries in these rapid, cell-surface initiated progesterone actions in endothelial and smooth muscle vascular cells. PRs have been detected in both cell types. New data are presented showing mPRα, mPRβ and mPRγ are also present in human endothelial and smooth muscle vascular cells. Preliminary evidence suggests mPRs mediate rapid progestin signaling in these endothelial cells, resulting in down-regulation of cAMP production and increased nitric oxide synthesis. The role of mPRs in progesterone regulation of cardiovascular functions warrants further investigation.
Collapse
Affiliation(s)
- Peter Thomas
- University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| | | |
Collapse
|
35
|
Kurakula K, Hamers AAJ, de Waard V, de Vries CJM. Nuclear Receptors in atherosclerosis: a superfamily with many 'Goodfellas'. Mol Cell Endocrinol 2013; 368:71-84. [PMID: 22664910 DOI: 10.1016/j.mce.2012.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/07/2023]
Abstract
Nuclear Receptors form a superfamily of 48 transcription factors that exhibit a plethora of functions in steroid hormone signaling, regulation of metabolism, circadian rhythm and cellular differentiation. In this review, we describe our current knowledge on the role of Nuclear Receptors in atherosclerosis, which is a multifactorial disease of the vessel wall. Various cell types are involved in this chronic inflammatory pathology in which multiple cellular processes and numerous genes are dysregulated. Systemic risk factors for atherosclerosis are among others adverse blood lipid profiles, enhanced circulating cytokine levels, as well as increased blood pressure. Since many Nuclear Receptors modulate lipid profiles or regulate blood pressure they indirectly affect atherosclerosis. In the present review, we focus on the functional involvement of Nuclear Receptors within the atherosclerotic vessel wall, more specifically on their modulation of cellular functions in endothelial cells, smooth muscle cells and macrophages. Collectively, this overview shows that most of the Nuclear Receptors are athero-protective in atherosclerotic lesions.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
36
|
Ribeiro RF, Potratz FF, Pavan BMM, Forechi L, Lima FLM, Fiorim J, Fernandes AA, Vassallo DV, Stefanon I. Carvedilol prevents ovariectomy-induced myocardial contractile dysfunction in female rat. PLoS One 2013; 8:e53226. [PMID: 23308166 PMCID: PMC3538779 DOI: 10.1371/journal.pone.0053226] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 11/26/2012] [Indexed: 11/18/2022] Open
Abstract
Carvedilol has beneficial effects on cardiac function in patients with heart failure but its effect on ovariectomy-induced myocardial contractile dysfunction remains unclear. Estrogen deficiency induces myocardial contractile dysfunction and increases cardiovascular disease risk in postmenopausal women. Our aim was to investigate whether carvedilol, a beta receptor blocker, would prevent ovariectomy-induced myocardial contractile dysfunction. Female rats (8 weeks old) that underwent bilateral ovariectomy were randomly assigned to receive daily treatment with carvedilol (OVX+CAR, 20 mg/kg), placebo (OVX) and SHAM for 58 days. Left ventricle papillary muscle was mounted for isometric tension recordings. The inotropic response to Ca2+ (0.62 to 3.75 mM) and isoproterenol (Iso 10−8 to 10−2 M) were assessed. Expression of calcium handling proteins was measured by western blot analysis. Carvedilol treatment in the OVX animals: prevented weight gain and slight hypertrophy, restored the reduced positive inotropic responses to Ca2+ and isoproterenol, prevented the reduction in SERCA2a expression, abolished the increase in superoxide anion production, normalized the increase in p22phox expression, and decreased serum angiotensin converting enzyme (ACE) activity. This study demonstrated that myocardial contractile dysfunction and SERCA2a down regulation were prevented by carvedilol treatment. Superoxide anion production and NADPH oxidase seem to be involved in this response.
Collapse
|
37
|
Reslan OM, Khalil RA. Vascular effects of estrogenic menopausal hormone therapy. Rev Recent Clin Trials 2012; 7:47-70. [PMID: 21864249 DOI: 10.2174/157488712799363253] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 07/22/2011] [Accepted: 07/29/2011] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) is more common in men and postmenopausal women (Post-MW) than premenopausal women (Pre-MW). Despite recent advances in preventive measures, the incidence of CVD in women has shown a rise that matched the increase in the Post-MW population. The increased incidence of CVD in Post-MW has been related to the decline in estrogen levels, and hence suggested vascular benefits of endogenous estrogen. Experimental studies have identified estrogen receptor ERα, ERβ and a novel estrogen binding membrane protein GPR30 (GPER) in blood vessels of humans and experimental animals. The interaction of estrogen with vascular ERs mediates both genomic and non-genomic effects. Estrogen promotes endothelium-dependent relaxation by increasing nitric oxide, prostacyclin, and hyperpolarizing factor. Estrogen also inhibits the mechanisms of vascular smooth muscle (VSM) contraction including [Ca2+]i, protein kinase C and Rho-kinase. Additional effects of estrogen on the vascular cytoskeleton, extracellular matrix, lipid profile and the vascular inflammatory response have been reported. In addition to the experimental evidence in animal models and vascular cells, initial observational studies in women using menopausal hormonal therapy (MHT) have suggested that estrogen may protect against CVD. However, randomized clinical trials (RCTs) such as the Heart and Estrogen/ progestin Replacement Study (HERS) and the Women's Health Initiative (WHI), which examined the effects of conjugated equine estrogens (CEE) in older women with established CVD (HERS) or without overt CVD (WHI), failed to demonstrate protective vascular effects of estrogen treatment. Despite the initial set-back from the results of MHT RCTs, growing evidence now supports the 'timing hypothesis', which suggests that MHT could increase the risk of CVD if started late after menopause, but may produce beneficial cardiovascular effects in younger women during the perimenopausal period. The choice of an appropriate MHT dose, route of administration, and estrogen/progestin combination could maximize the vascular benefits of MHT and minimize other adverse effects, especially if given within a reasonably short time after menopause to women that seek MHT for the relief of menopausal symptoms.
Collapse
Affiliation(s)
- Ossama M Reslan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | |
Collapse
|
38
|
Mann MC, Exner DV, Hemmelgarn BR, Turin TC, Sola DY, Ahmed SB. Impact of gender on the cardiac autonomic response to angiotensin II in healthy humans. J Appl Physiol (1985) 2012; 112:1001-7. [DOI: 10.1152/japplphysiol.01207.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Premenopausal women have a lower risk of cardiovascular disease (CVD) compared with men of a similar age. Furthermore, the regulation of factors that influence CVD appears to differ between the sexes, including control of the autonomic nervous system (ANS) and the renin-angiotensin system. We examined the cardiac ANS response to angiotensin II (Ang II) challenge in healthy subjects to determine whether differences in women and men exist. Thirty-six healthy subjects (21 women, 15 men, age 38 ± 2 years) were studied in a high-salt balance. Heart-rate variability (HRV) was calculated by spectral power analysis [low-frequency (LF) sympathetic modulation, high-frequency (HF) parasympathetic/vagal modulation, and LF:HF as a measure of overall ANS balance]. HRV was assessed at baseline and in response to graded Ang II infusions (3 ng·kg−1·min−1 × 30 min; 6 ng·kg−1·min−1 × 30 min). Cardiac ANS tone did not change significantly in women after each Ang II dose [3 ng·kg−1·min−1 mean change (Δ)LF:HF (mean ± SE) 0.5 ± 0.3, P = 0.8, vs. baseline; 6 ng·kg−1·min−1 ΔLF:HF (mean ± SE) 0.5 ± 0.4, P = 0.4, vs. baseline], whereas men exhibited an unfavorable shift in overall cardiac ANS activity in response to Ang II (ΔLF:HF 2.6 ± 0.2, P = 0.01, vs. baseline; P = 0.02 vs. female response). This imbalance in sympathovagal tone appeared to be largely driven by a withdrawal in cardioprotective vagal activity in response to Ang II challenge [ΔHF normalized units (nu), −5.8 ± 2.9, P = 0.01, vs. baseline; P = 0.006 vs. women] rather than an increase in sympathetic activity (ΔLF nu, −4.5 ± 5.7, P = 0.3, vs. baseline; P = 0.5 vs. women). Premenopausal women maintain cardiac ANS tone in response to Ang II challenge, whereas similarly aged men exhibit an unfavorable shift in cardiovagal activity. Understanding the role of gender in ANS modulation may help guide risk-reduction strategies in high-risk CVD populations.
Collapse
Affiliation(s)
- M. C. Mann
- Faculty of Medicine, University of Calgary
- Libin Cardiovascular Institute of Alberta, Calgary; and
| | - D. V. Exner
- Faculty of Medicine, University of Calgary
- Libin Cardiovascular Institute of Alberta, Calgary; and
| | - B. R. Hemmelgarn
- Faculty of Medicine, University of Calgary
- Libin Cardiovascular Institute of Alberta, Calgary; and
- Alberta Kidney Disease Network, Alberta, Canada
| | | | - D. Y. Sola
- Libin Cardiovascular Institute of Alberta, Calgary; and
| | - S. B. Ahmed
- Faculty of Medicine, University of Calgary
- Libin Cardiovascular Institute of Alberta, Calgary; and
- Alberta Kidney Disease Network, Alberta, Canada
| |
Collapse
|
39
|
Yasuda N, Akazawa H, Ito K, Shimizu I, Kudo-Sakamoto Y, Yabumoto C, Yano M, Yamamoto R, Ozasa Y, Minamino T, Naito AT, Oka T, Shiojima I, Tamura K, Umemura S, Paradis P, Nemer M, Komuro I. Agonist-Independent Constitutive Activity of Angiotensin II Receptor Promotes Cardiac Remodeling in Mice. Hypertension 2012; 59:627-33. [DOI: 10.1161/hypertensionaha.111.175208] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The angiotensin II (Ang II) type 1 (AT
1
) receptor mainly mediates the physiological and pathological actions of Ang II, but recent studies have suggested that AT
1
receptor inherently shows spontaneous constitutive activity even in the absence of Ang II in culture cells. To elucidate the role of Ang II–independent AT
1
receptor activation in the pathogenesis of cardiac remodeling, we generated transgenic mice overexpressing AT
1
receptor under the control of α-myosin heavy chain promoter in angiotensinogen-knockout background (AT
1
Tg-AgtKO mice). In AT
1
Tg-AgtKO hearts, redistributions of the Gα
q11
subunit into cytosol and phosphorylation of extracellular signal-regulated kinases were significantly increased, compared with angiotensinogen-knockout mice hearts, suggesting that the AT
1
receptor is constitutively activated independent of Ang II. As a consequence, AT
1
Tg-AgtKO mice showed spontaneous systolic dysfunction and chamber dilatation, accompanied by severe interstitial fibrosis. Progression of cardiac remodeling in AT
1
Tg-AgtKO mice was prevented by treatment with candesartan, an inverse agonist for the AT
1
receptor, but not by its derivative candesartan-7H, deficient of inverse agonism attributed to a lack of the carboxyl group at the benzimidazole ring. Our results demonstrate that constitutive activity of the AT
1
receptor under basal conditions contributes to the cardiac remodeling even in the absence of Ang II, when the AT
1
receptor is upregulated in the heart.
Collapse
Affiliation(s)
- Noritaka Yasuda
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Hiroshi Akazawa
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Kaoru Ito
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Ippei Shimizu
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Yoko Kudo-Sakamoto
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Chizuru Yabumoto
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Masamichi Yano
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Rie Yamamoto
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Yukako Ozasa
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Tohru Minamino
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Atsuhiko T. Naito
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Toru Oka
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Ichiro Shiojima
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Kouichi Tamura
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Satoshi Umemura
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Pierre Paradis
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Mona Nemer
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| | - Issei Komuro
- From the Department of Cardiovascular Science and Medicine (N.Y., K.I., Ip.S., R.Y., Y.O., T.M.), Chiba University Graduate School of Medicine, Chiba, Japan; Departments of Cardiovascular Medicine (H.A, Y.K.-S., C.Y., M.Y., T.O., I.K.) and Cardiovascular Regenerative Medicine (A.T.N., Ic.S.), Osaka University Graduate School of Medicine, Suita, Japan; Department of Medical Science and Cardiorenal Medicine (K.T., S.U.), Yokohama City University Graduate School of Medicine, Yokohama, Japan; Lady Davis
| |
Collapse
|
40
|
Sampson AK, Hilliard LM, Moritz KM, Thomas MC, Tikellis C, Widdop RE, Denton KM. The arterial depressor response to chronic low-dose angiotensin II infusion in female rats is estrogen dependent. Am J Physiol Regul Integr Comp Physiol 2011; 302:R159-65. [PMID: 22031787 DOI: 10.1152/ajpregu.00256.2011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The complex role of the renin-angiotensin-system (RAS) in arterial pressure regulation has been well documented. Recently, we demonstrated that chronic low-dose angiotensin II (ANG II) infusion decreases arterial pressure in female rats via an AT(2)R-mediated mechanism. Estrogen can differentially regulate components of the RAS and is known to influence arterial pressure regulation. We hypothesized that AT(2)R-mediated depressor effects evident in females were estrogen dependent and thus would be abolished by ovariectomy and restored by estrogen replacement. Female Sprague-Dawley rats underwent ovariectomy or sham surgery and were treated with 17β-estradiol or placebo. Mean arterial pressure (MAP) was measured via telemetry in response to a 2-wk infusion of ANG II (50 ng·kg(-1)·min(-1) sc) or saline. MAP significantly decreased in females treated with ANG II (-10 ± 2 mmHg), a response that was abolished by ovariectomy (+4 ± 2 mmHg) and restored with estrogen replacement (-6 ± 2 mmHg). Cardiac and renal gene expression of components of the RAS was differentially regulated by estrogen, such that overall, estrogen shifted the balance of the RAS toward the vasodilatory axis. In conclusion, estrogen-dependent mechanisms offset the vasopressor actions of ANG II by enhancing RAS vasodilator pathways in females. This highlights the potential for these vasodilator pathways as therapeutic targets, particularly in women.
Collapse
Affiliation(s)
- Amanda K Sampson
- Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
41
|
Ross RL, Serock MR, Khalil RA. Experimental benefits of sex hormones on vascular function and the outcome of hormone therapy in cardiovascular disease. Curr Cardiol Rev 2011; 4:309-22. [PMID: 20066139 PMCID: PMC2801863 DOI: 10.2174/157340308786349462] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 06/16/2008] [Accepted: 06/16/2008] [Indexed: 11/22/2022] Open
Abstract
Cardiovascular disease (CVD) is more common in men and postmenopausal women than premenopausal women, suggesting vascular benefits of female sex hormones. Experimental data have shown beneficial vascular effects of estrogen including stimulation of endothelium-dependent nitric oxide, prostacyclin and hyperpolarizing factor-mediated vascular relaxation. However, the experimental evidence did not translate into vascular benefits of hormone replacement therapy (HRT) in postmenopausal women, and HERS, HERS-II and WHI clinical trials demonstrated adverse cardiovascular events with HRT. The lack of vascular benefits of HRT could be related to the hormone used, the vascular estrogen receptor (ER), and the subject’s age and preexisting cardiovascular condition. Natural and phytoestrogens in small doses may be more beneficial than synthetic estrogen. Specific estrogen receptor modulators (SERMs) could maximize the vascular benefits, with little side effects on breast cancer. Transdermal estrogens avoid the first-pass liver metabolism associated with the oral route. Postmenopausal decrease and genetic polymorphism in vascular ER and post-receptor signaling mechanisms could also modify the effects of HRT. Variants of cytosolic/nuclear ER mediate transcriptional genomic effects that stimulate endothelial cell growth, but inhibit vascular smooth muscle (VSM) proliferation. Also, plasma membrane ERs trigger not only non-genomic stimulation of endothelium-dependent vascular relaxation, but also inhibition of [Ca2+]i, protein kinase C and Rho kinase-dependent VSM contraction. HRT could also be more effective in the perimenopausal period than in older postmenopausal women, and may prevent the development, while worsening preexisting CVD. Lastly, progesterone may modify the vascular effects of estrogen, and modulators of estrogen/testosterone ratio could provide alternative HRT combinations. Thus, the type, dose, route of administration and the timing/duration of HRT should be customized depending on the subject’s age and preexisting cardiovascular condition, and thereby make it possible to translate the beneficial vascular effects of sex hormones to the outcome of HRT in postmenopausal CVD.
Collapse
Affiliation(s)
- Reagan L Ross
- Division of Vascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
42
|
Schober J, Weil Z, Pfaff D. How generalized CNS arousal strengthens sexual arousal (and vice versa). Horm Behav 2011; 59:689-95. [PMID: 20950622 DOI: 10.1016/j.yhbeh.2010.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 09/23/2010] [Accepted: 10/02/2010] [Indexed: 12/22/2022]
Abstract
Heightened states of generalized CNS arousal are proposed here to facilitate sexual arousal in both males and females. Genetic, pharmacologic and biophysical mechanisms by which this happens are reviewed. Moreover, stimulation of the genital epithelia, as triggers of sex behavior, is hypothesized to lead to a greater generalized arousal in a manner that intensifies sexual motivation. Finally, launched from histochemical studies intended to characterize cells in the genital epithelium, a surprising idea is proposed that links density of innervation with the efficiency of wound healing and with the capacity of that epithelium to stimulate generalized CNS arousal. Thus, bidirectional arousal-related mechanisms that foster sexual behaviors are envisioned as follows: from specific to generalized (as with genital stimulation) and from generalized to specific.
Collapse
Affiliation(s)
- Justine Schober
- Laboratory of Neurobiology and Behavior, The Rockefeller University, NY, USA
| | | | | |
Collapse
|
43
|
Wu Q, Chambliss K, Umetani M, Mineo C, Shaul PW. Non-nuclear estrogen receptor signaling in the endothelium. J Biol Chem 2011; 286:14737-43. [PMID: 21343284 PMCID: PMC3083154 DOI: 10.1074/jbc.r110.191791] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In addition to the classical function of estrogen receptors (ER) as transcription factors, evidence continues to accumulate that they mediate non-nuclear processes in numerous cell types, including the endothelium, in which they activate endothelial NO synthase. Non-nuclear ER signaling entails unique post-translational modifications and protein-protein interactions of the receptor with adaptor molecules, kinases, and G proteins. Recent in vitro and in vivo studies in mice using an estrogen-dendrimer conjugate that is excluded from the nucleus indicate that non-nuclear ER activation underlies the migration and growth responses of endothelial cells to estrogen but not the growth responses of endometrial or breast cancer cells to the hormone. In this minireview, the features of ERα and protein-protein interactions that enable it to invoke extranuclear signaling in the endothelium and the consequences of that signaling are discussed.
Collapse
Affiliation(s)
- Qian Wu
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ken Chambliss
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Michihisa Umetani
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Chieko Mineo
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Philip W. Shaul
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
44
|
Masood DEN, Roach EC, Beauregard KG, Khalil RA. Impact of sex hormone metabolism on the vascular effects of menopausal hormone therapy in cardiovascular disease. Curr Drug Metab 2011; 11:693-714. [PMID: 21189141 DOI: 10.2174/138920010794233477] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 10/25/2010] [Indexed: 12/24/2022]
Abstract
Epidemiological studies have shown that cardiovascular disease (CVD) is less common in pre-menopausal women (Pre-MW) compared to men of the same age or post-menopausal women (Post-MW), suggesting cardiovascular benefits of estrogen. Estrogen receptors (ERs) have been identified in the vasculature, and experimental studies have demonstrated vasodilator effects of estrogen/ER on the endothelium, vascular smooth muscle (VSM) and extracellular matrix. Several natural and synthetic estrogenic preparations have been developed for relief of menopausal vasomotor symptoms. However, whether menopausal hormone therapy (MHT) is beneficial in postmenopausal CVD remains controversial. Despite reports of vascular benefits of MHT from observational and experimental studies, randomized clinical trials (RCTs), such as the Heart and Estrogen/progestin Replacement Study (HERS) and the Women's Health Initiative (WHI), have suggested that, contrary to expectations, MHT may increase the risk of CVD. These discrepancies could be due to agerelated changes in sex hormone synthesis and metabolism, which would influence the effective dose of MHT and the sex hormone environment in Post-MW. Age-related changes in the vascular ER subtype, structure, expression, distribution, and post-ER signaling pathways in the endothelium and VSM, along with factors related to the design of RCTs, preexisting CVD condition, and structural changes in the blood vessels architecture have also been suggested as possible causes of MHT failure in CVD. Careful examination of these factors should help in identifying the causes of the changes in the vascular effects of estrogen with age. The sex hormone metabolic pathways, the active versus inactive estrogen metabolites, and their effects on vascular function, the mitochondria, the inflammatory process and angiogenesis should be further examined. Also, the genomic and non-genomic effects of estrogenic compounds should be viewed as integrated rather than discrete responses. The complex interactions between these factors highlight the importance of careful design of MHT RCTs, and the need of a more customized approach for each individual patient in order to enhance the vascular benefits of MHT in postmenopausal CVD.
Collapse
Affiliation(s)
- Durr-e-Nayab Masood
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
45
|
Komukai K, Mochizuki S, Yoshimura M. Gender and the renin-angiotensin-aldosterone system. Fundam Clin Pharmacol 2011; 24:687-98. [PMID: 20608988 DOI: 10.1111/j.1472-8206.2010.00854.x] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Premenopausal women are protected to some extent from cardiovascular and kidney diseases. Because this protection weakens after menopause, sex hormones are believed to play an important role in the pathogenesis of cardiovascular and kidney diseases. The cardiovascular system and the kidneys are regulated by the renin-angiotensin-aldosterone system (RAAS), which in turn, appears to be regulated by sex hormones. In general, oestrogen increases angiotensinogen levels and decreases renin levels, angiotensin-converting enzyme (ACE) activity, AT(1) receptor density, and aldosterone production. Oestrogen also activates counterparts of the RAAS such as natriuretic peptides, AT(2) receptor density, and angiotensinogen (1-7). Progesterone competes with aldosterone for mineralocorticoid receptor. Less is known about androgens, but testosterone seems to increase renin levels and ACE activity. These effects of sex hormones on the RAAS can explain at least some of the gender differences in cardiovascular and kidney diseases.
Collapse
Affiliation(s)
- Kimiaki Komukai
- Division of Cardiology, The Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | | | | |
Collapse
|
46
|
Morrissy S, Xu B, Aguilar D, Zhang J, Chen QM. Inhibition of apoptosis by progesterone in cardiomyocytes. Aging Cell 2010; 9:799-809. [PMID: 20726854 DOI: 10.1111/j.1474-9726.2010.00619.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
While gender-based differences in heart disease have raised the possibility that estrogen (ES) or progesterone (PG) may have cardioprotective effects, recent controversy regarding hormone replacement therapy has questioned the cardiac effects of these steroids. Using cardiomyocytes, we tested whether ES or PG has protective effects at the cellular level. We found that PG but not ES protects cardiomyocytes from apoptotic cell death induced by doxorubicin (Dox). PG inhibited apoptosis in a dose-dependent manner, by 12 ± 4.0% at 1 μm and 60 ± 1.0% at 10 μm. The anti-apoptotic effect of PG was also time dependent, causing 18 ± 5% or 62 + 2% decrease in caspase-3 activity within 1 h or 72 h of pretreatment. While PG causes nuclear translocation of its receptor within 20 min, the cytoprotective effect of PG was canceled by mifepristone (MF), a PG receptor antagonist. Analyses using Affymetrix high-density oligonucleotide array and RT-PCR found that PG induced Bcl-xL, metallothionine, NADPH quinone oxidoreductase 1, glutathione peroxidase-3, and four isoforms of glutathione S-transferase. Western blot analyses revealed that PG indeed induced an elevation of Bcl-xL protein in a dose- and time-dependent manner. Nuclear run-on assay indicated that PG induced Bcl-xL gene transcription. Inhibiting the expression of Bcl-xL using siRNA reduced the cytoprotective effect of PG. Our data suggests that PG induces a cytoprotective effect in cardiomyocytes in association with induction of Bcl-xL gene.
Collapse
Affiliation(s)
- Stephen Morrissy
- Department of Pharmacology, University of Arizona, Tucson, 85724, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
CVD (cardiovascular disease) is the leading cause of death for women. Considerable progress has been made in both our understanding of the complexities governing menopausal hormone therapy and our understanding of the cellular and molecular mechanisms underlying hormone and hormone receptor function. Understanding the interplay of atherosclerosis and sex steroid hormones and their cognate receptors at the level of the vessel wall has important ramifications for clinical practice. In the present review, we discuss the epidemiology of CVD in men and women, the clinical impact of sex hormones on CVD, and summarize our current understanding of the pathogenesis of atherosclerosis with a focus on gender differences in CVD, its clinical presentation and course, and pathobiology. The critical animal and human data that pertain to the role of oestrogens, androgens and progestins on the vessel wall is also reviewed, with particular attention to the actions of sex hormones on each of the three key cell types involved in atherogenesis: the endothelium, smooth muscle cells and macrophages. Where relevant, the systemic (metabolic) effects of sex hormones that influence atherogenesis, such as those involving vascular reactivity, inflammation and lipoprotein metabolism, are discussed. In addition, four key current concepts in the field are explored: (i) total hormone exposure time and coronary heart disease risk; (ii) the importance of tissue specificity of sex steroid hormones, critical timing and the stage of atherosclerosis in hormone action; (iii) biomarkers for atherosclerosis with regard to hormone therapy; and (iv) the complex role of sex steroids in inflammation. Future studies in this field will contribute to guiding clinical treatment recommendations for women and help define research priorities.
Collapse
|
48
|
Castrop H, Höcherl K, Kurtz A, Schweda F, Todorov V, Wagner C. Physiology of Kidney Renin. Physiol Rev 2010; 90:607-73. [PMID: 20393195 DOI: 10.1152/physrev.00011.2009] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The protease renin is the key enzyme of the renin-angiotensin-aldosterone cascade, which is relevant under both physiological and pathophysiological settings. The kidney is the only organ capable of releasing enzymatically active renin. Although the characteristic juxtaglomerular position is the best known site of renin generation, renin-producing cells in the kidney can vary in number and localization. (Pro)renin gene transcription in these cells is controlled by a number of transcription factors, among which CREB is the best characterized. Pro-renin is stored in vesicles, activated to renin, and then released upon demand. The release of renin is under the control of the cAMP (stimulatory) and Ca2+(inhibitory) signaling pathways. Meanwhile, a great number of intrarenally generated or systemically acting factors have been identified that control the renin secretion directly at the level of renin-producing cells, by activating either of the signaling pathways mentioned above. The broad spectrum of biological actions of (pro)renin is mediated by receptors for (pro)renin, angiotensin II and angiotensin-( 1 – 7 ).
Collapse
Affiliation(s)
- Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Klaus Höcherl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Vladimir Todorov
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
49
|
Sakuta T, Morita Y, Satoh M, Fox DA, Kashihara N. Involvement of the renin-angiotensin system in the development of vascular damage in a rat model of arthritis: Effect of angiotensin receptor blockers. ACTA ACUST UNITED AC 2010; 62:1319-28. [DOI: 10.1002/art.27384] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
do Nascimento GRA, Barros YVR, Wells AK, Khalil RA. Research into Specific Modulators of Vascular Sex Hormone Receptors in the Management of Postmenopausal Cardiovascular Disease. Curr Hypertens Rev 2009; 5:283-306. [PMID: 20694192 PMCID: PMC2915874 DOI: 10.2174/157340209789587717] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cardiovascular disease (CVD) is more common in men and postmenopausal women than premenopausal women, suggesting vascular benefits of female sex hormones. Studies on the vasculature have identified estrogen receptors ERα, ERβ and a novel estrogen binding membrane protein GPR30, that mediate genomic and/or non-genomic effects. Estrogen promotes endothelium-dependent relaxation by inducing the production/activity of nitric oxide, prostacyclin, and hyperpolarizing factor, and inhibits the mechanisms of vascular smooth muscle contraction including [Ca(2+)](i), protein kinase C, Rho kinase and mitogen-activated protein kinase. Additional effects of estrogen on the cytoskeleton, matrix metalloproteinases and inflammatory factors contribute to vascular remodeling. However, the experimental evidence did not translate into vascular benefits of menopausal hormone therapy (MHT), and the HERS, HERS-II and WHI clinical trials demonstrated adverse cardiovascular events. The discrepancy has been partly related to delayed MHT and potential changes in the vascular ER amount, integrity, affinity, and downstream signaling pathways due to the subjects' age and preexisting CVD. The adverse vascular effects of MHT also highlighted the need of specific modulators of vascular sex hormone receptors. The effectiveness of MHT can be improved by delineating the differences in phramcokinetics and pharmacodynamics of natural, synthetic, and conjugated equine estrogens. Estriol, "hormone bioidenticals" and phytoestrogens are potential estradiol substitutes. The benefits of low dose MHT, and transdermal or vaginal estrogens over oral preparations are being evaluated. Specific ER modulators (SERMs) and ER agonists are being developed to maximize the effects on vascular ERs. Also, the effects of estrogen are being examined in the context of the whole body hormonal environment and the levels of progesterone and androgens. Thus, the experimental vascular benefits of estrogen can be translated to the outcome of MHT in postmenopausal CVD, as more specific modulators of sex hormone receptors become available and are used at the right dose, route of administration and timing, depending on the subject's age and preexisting cardiovascular condition.
Collapse
|