1
|
Pérez-Coria M, Vázquez-Rivera GE, Gómez-García EF, Mendoza-Carrera F. Sex differences in fetal kidney reprogramming: the case in the renin-angiotensin system. Pediatr Nephrol 2024; 39:645-653. [PMID: 37572115 DOI: 10.1007/s00467-023-06112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/14/2023]
Abstract
During the early stages of the development of the living multiorgan systems, genome modifications other than sequence variation occur that guide cell differentiation and organogenesis. These modifications are known to operate as a fetal programming code during this period, and recent research indicates that there are some tissue-specific codes in organogenesis whose effects may persist after birth until adulthood. Consequently, the events that disrupt the pre-established epigenetic pattern could induce shifts in organ physiology, with implications on health from birth or later in adult life. Chronic kidney disease (CKD) is one of the main causes of mortality worldwide; its etiology is multifactorial, but diabetes, obesity, and hypertension are the main causes of CKD in adults, although there are other risk factors that are mainly associated with an individual's lifestyle. Recent studies suggest that fetal reprogramming in the developing kidney could be implicated in the susceptibility to kidney disease in both childhood and adulthood. Some epigenetic modifications, such as genome methylation status, dysregulation of miRNA, and histone coding alterations in genes related to the regulation of the renin-angiotensin axis, a common denominator in CKD, may have originated during fetal development. This review focuses on epigenetic changes during nephrogenesis and their repercussions on kidney health and disease. In addition, the focus is on the influence of environmental factors during pregnancy, such as maternal metabolic diseases and dietary and metabolic conditions, as well as some sex differences in fetal kidney reprogramming during which dysregulation of the renin-angiotensin system is involved.
Collapse
Affiliation(s)
- Mariana Pérez-Coria
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Gloria Elizabeth Vázquez-Rivera
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Erika Fabiola Gómez-García
- Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Tijuana, Baja California, Mexico
| | - Francisco Mendoza-Carrera
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
2
|
Cheng HL, Garden FL, Skilton MR, Johnson C, Webster J, Grimes CA, Ivers RQ, Steinbeck KS. Impact of growth, gonadal hormones, adiposity and the sodium-to-potassium ratio on longitudinal adolescent measures of blood pressure at puberty. J Hum Hypertens 2023; 37:835-843. [PMID: 36376566 DOI: 10.1038/s41371-022-00774-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022]
Abstract
Blood pressure (BP) rises rapidly at puberty. While this is partly due to normal development, factors like excess adiposity and a high intake of dietary sodium relative to potassium may contribute to a true increase in hypertension risk. This study aimed to assess the relative impact of growth, gonadal hormones, adiposity and the sodium-to-potassium ratio (Na:K) on longitudinal BP measures at puberty. This study analysed data from a three-year longitudinal cohort study of pubertal adolescents. Anthropometry, body composition (bio-electrical impedance), serum testosterone and oestradiol (mass spectrometry) were measured annually. Na:K was measured from three-monthly urine samples. These variables were used to predict annual BP measures using mixed modelling and ordinal regression. Data from 325 adolescents (11.7 ± 1.0 y; 55% male) were analysed, showing typical growth patterns at puberty. Systolic BP increased over time in both sexes (p < 0.01), with boys exhibiting a significantly steeper rise compared to girls. Adiposity variables (BMI z-score, percent body fat, fat mass, waist-to-height ratio) strongly and consistently predicted systolic and diastolic BP in both sexes (all p < 0.05). Systolic BP was also significantly and positively related to height (p < 0.05). No associations with BP were identified in either sex for gonadal hormones or Na:K. Similar results were obtained when BP was classified into hypertension categories. Relative to other developmental and diet-related variables tested, adiposity was found to be the strongest most consistent predictor of BP in pubertal adolescents. Findings highlight the importance of dedicated youth obesity management interventions and policy measures for reducing long-term hypertension and cardiovascular disease risks.Australian New Zealand Clinical Trials Registry ACTRN12617000964314.
Collapse
Affiliation(s)
- Hoi Lun Cheng
- The Children's Hospital at Westmead, Academic Department of Adolescent Medicine, Westmead, NSW, Australia.
- The University of Sydney, Faculty of Medicine and Health, Sydney Medical School, Specialty of Child and Adolescent Health, Westmead, NSW, Australia.
- The University of Sydney, Faculty of Medicine and Health, The Boden Initiative, Charles Perkins Centre, Camperdown, NSW, Australia.
| | - Frances L Garden
- University of New South Wales, School of Clinical Medicine, South West Sydney Clinical Campuses, Liverpool, NSW, Australia
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| | - Michael R Skilton
- The University of Sydney, Faculty of Medicine and Health, The Boden Initiative, Charles Perkins Centre, Camperdown, NSW, Australia
| | - Claire Johnson
- University of New South Wales, The George Institute for Global Health, Sydney, NSW, Australia
- University of New South Wales, School of Population Health, Sydney, NSW, Australia
| | - Jacqui Webster
- University of New South Wales, The George Institute for Global Health, Sydney, NSW, Australia
- University of New South Wales, School of Population Health, Sydney, NSW, Australia
| | - Carley A Grimes
- Deakin University, Institute for Physical Activity and Nutrition, Geelong, VIC, Australia
| | - Rebecca Q Ivers
- University of New South Wales, The George Institute for Global Health, Sydney, NSW, Australia
- University of New South Wales, School of Population Health, Sydney, NSW, Australia
| | - Katharine S Steinbeck
- The Children's Hospital at Westmead, Academic Department of Adolescent Medicine, Westmead, NSW, Australia
- The University of Sydney, Faculty of Medicine and Health, Sydney Medical School, Specialty of Child and Adolescent Health, Westmead, NSW, Australia
- The University of Sydney, Faculty of Medicine and Health, The Boden Initiative, Charles Perkins Centre, Camperdown, NSW, Australia
| |
Collapse
|
3
|
Hall E, Vrolijk MF. Androgen Receptor and Cardiovascular Disease: A Potential Risk for the Abuse of Supplements Containing Selective Androgen Receptor Modulators. Nutrients 2023; 15:3330. [PMID: 37571268 PMCID: PMC10420890 DOI: 10.3390/nu15153330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The androgen receptor (AR) is a member of the family of ligand-activated transcription factors. Selective androgen receptor modulators (SARMs) exert their biological function through complex interactions with the AR. It has been speculated that overexertion of AR signaling cascades as a result of SARM abuse can be a risk factor for the development of various cardiovascular diseases. The present literature review explores the implications of the interaction between SARMs and the AR on cardiovascular health by focusing on the AR structure, function, and mechanisms of action, as well as the current clinical literature on various SARMs. It is shown that SARMs may increase the risk of cardiovascular diseases through implications on the renin-angiotensin system, smooth muscle cells, sympathetic nervous system, lipid profile, inflammation, platelet activity, and various other factors. More research on this topic is necessary as SARM abuse is becoming increasingly common. There is a noticeable lack of clinical trials and literature on the relationship between SARMs, cardiovascular diseases, and the AR. Future in vivo and in vitro studies within this field are vital to understand the mechanisms that underpin these complex interactions and risk factors.
Collapse
Affiliation(s)
| | - Misha F. Vrolijk
- Department of Pharmacology and Toxicology, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
4
|
Gong S, Sun N, Meyer LS, Tetti M, Koupourtidou C, Krebs S, Masserdotti G, Blum H, Rainey WE, Reincke M, Walch A, Williams TA. Primary Aldosteronism: Spatial Multiomics Mapping of Genotype-Dependent Heterogeneity and Tumor Expansion of Aldosterone-Producing Adenomas. Hypertension 2023; 80:1555-1567. [PMID: 37125608 PMCID: PMC10330203 DOI: 10.1161/hypertensionaha.123.20921] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/10/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Primary aldosteronism is frequently caused by an adrenocortical aldosterone-producing adenoma (APA) carrying a somatic mutation that drives aldosterone overproduction. APAs with a mutation in KCNJ5 (APA-KCNJ5MUT) are characterized by heterogeneous CYP11B2 (aldosterone synthase) expression, a particular cellular composition and larger tumor diameter than those with wild-type KCNJ5 (APA-KCNJ5WT). We exploited these differences to decipher the roles of transcriptome and metabolome reprogramming in tumor pathogenesis. METHODS Consecutive adrenal cryosections (7 APAs and 7 paired adjacent adrenal cortex) were analyzed by spatial transcriptomics (10x Genomics platform) and metabolomics (in situ matrix-assisted laser desorption/ionization mass spectrometry imaging) co-integrated with CYP11B2 immunohistochemistry. RESULTS We identified intratumoral transcriptional heterogeneity that delineated functionally distinct biological pathways. Common transcriptomic signatures were established across all APA specimens which encompassed 2 distinct transcriptional profiles in CYP11B2-immunopositive regions (CYP11B2-type 1 or 2). The CYP11B2-type 1 signature was characterized by zona glomerulosa gene markers and was detected in both APA-KCNJ5MUT and APA-KCNJ5WT. The CYP11B2-type 2 signature displayed markers of the zona fasciculata or reticularis and predominated in APA-KCNJ5MUT. Metabolites that promote oxidative stress and cell death accumulated in APA-KCNJ5WT. In contrast, antioxidant metabolites were abundant in APA-KCNJ5MUT. Finally, APA-like cell subpopulations-negative for CYP11B2 gene expression-were identified in adrenocortical tissue adjacent to APAs suggesting the existence of tumor precursor states. CONCLUSIONS Our findings provide insight into intra- and intertumoral transcriptional heterogeneity and support a role for prooxidant versus antioxidant systems in APA pathogenesis highlighting genotype-dependent capacities for tumor expansion.
Collapse
Affiliation(s)
- Siyuan Gong
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Na Sun
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Germany
| | - Lucie S Meyer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Martina Tetti
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Christina Koupourtidou
- Department for Cell Biology and Anatomy, Biomedical Center, Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
- Graduate School Systemic Neurosciences, Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, 81377 Munich, Germany
| | - Giacomo Masserdotti
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, 81377 Munich, Germany
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Metabolism, Endocrine, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum München, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| |
Collapse
|
5
|
Yanes Cardozo LL, Romero DG. Management of cardiometabolic complications in polycystic ovary syndrome: Unmet needs. FASEB J 2021; 35:e21945. [PMID: 34606638 PMCID: PMC10146586 DOI: 10.1096/fj.202002526rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 08/05/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder and the most common cause of androgen excess in reproductive-age women. The heterogeneity of the clinical presentation in PCOS patients suggests the involvement of multiples abnormal physiological pathways. In addition, women with PCOS have a high prevalence of cardiometabolic risk factors. Unfortunately, limited effective evidence-based therapeutic agents are available to treat the cardiometabolic complications in PCOS patients. Insights from recent studies highlight the multiple opportunities to deliver timely effective medical care for women with PCOS. This perspective manuscript aims to highlight the unmet need for effective and safe management of the cardiometabolic complications in PCOS patients.
Collapse
Affiliation(s)
- Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
6
|
The Sexually Dimorphic Adrenal Cortex: Implications for Adrenal Disease. Int J Mol Sci 2021; 22:ijms22094889. [PMID: 34063067 PMCID: PMC8124132 DOI: 10.3390/ijms22094889] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Many adrenocortical diseases are more prevalent in women than in men, but the reasons underlying this sex bias are still unknown. Recent studies involving gonadectomy and sex hormone replacement experiments in mice have shed some light onto the molecular basis of sexual dimorphism in the adrenal cortex. Indeed, it has been shown that gonadal hormones influence many aspects of adrenal physiology, ranging from stem cell-dependent tissue turnover to steroidogenesis and X-zone dynamics. This article reviews current knowledge on adrenal cortex sexual dimorphism and the potential mechanisms underlying sex hormone influence of adrenal homeostasis. Both topics are expected to contribute to personalized and novel therapeutic approaches in the future.
Collapse
|
7
|
Gao X, Yamazaki Y, Tezuka Y, Omata K, Ono Y, Morimoto R, Nakamura Y, Satoh F, Sasano H. Gender differences in human adrenal cortex and its disorders. Mol Cell Endocrinol 2021; 526:111177. [PMID: 33582213 DOI: 10.1016/j.mce.2021.111177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
The adrenal cortex plays pivotal roles in the maintenance of blood volume, responsiveness to stress and the development of gender characteristics. Gender differences of human adrenal cortex have been recently reported and attracted increasing interests. Gender differences occur from the developing stage of the adrenal, in which female subjects had more activated stem cells with higher renewal capacity resulting in gender-associated divergent structures and functions of cortical zonations of human adrenal. Female subjects generally have the lower blood pressure with the lower renin levels and ACE activities than male subjects. In addition, HPA axis was more activated in female than male, which could possibly contribute to gender differences in coping with various stressful events in our life. Of particular interest, estrogens were reported to suppress RAAS but activate HPA axis, whereas androgens had opposite effects. In addition, adrenocortical disorders in general occur more frequently in female with more pronounced adrenocortical hormonal abnormalities possibly due to their more activated WNT and PRK signaling pathways with more abundant activated adrenocortical stem cells present in female adrenal glands. Therefore, it has become pivotal to clarify the gender influence on both clinical and biological features of adrenocortical disorders. We herein reviewed recent advances in these fields.
Collapse
Affiliation(s)
- Xin Gao
- Department of Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Yuta Tezuka
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Japan
| | - Kei Omata
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Japan
| | - Yoshikiyo Ono
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Japan
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Japan
| | - Fumitoshi Satoh
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Japan.
| |
Collapse
|
8
|
Salvador AC, Arends D, Barrington WT, Elsaadi AM, Brockmann GA, Threadgill DW. Sex-specific genetic architecture in response to American and ketogenic diets. Int J Obes (Lond) 2021; 45:1284-1297. [PMID: 33723359 PMCID: PMC8159743 DOI: 10.1038/s41366-021-00785-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/19/2020] [Accepted: 02/02/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND/OBJECTIVES There is a growing appreciation for individual responses to diet. In a previous study, mouse strain-specific responses to American and ketogenic diets were observed. In this study, we searched for genetic variants underlying differences in the responses to American and ketogenic diets between C57BL/6J (B6) and FVB/NJ (FVB) mouse strains. RESULTS Genetic mapping of fat and lean mass gain revealed QTLs on Chromosome (Chr) 1 at 191.6 Mb (Fmgq1) (P < 0.001, CI = 180.2-194.4 Mb), Chr5 at 73.7 Mb (Fmgq2, Lmgq1) (P < 0.001, CI = 66.1-76.6 Mb), and Chr7 at 40.5 Mb (Fmgq3) (P < 0.01, CI = 36.6-44.5 Mb). Analysis of serum HDL cholesterol concentration identified a significant (P < 0.001, CI = 160.6-176.1 Mb) QTL on Chr1 at 168.6 Mb (Hdlq1). Causal network inference suggests that HDL cholesterol and fat mass gain are both linked to Fmgq1. CONCLUSIONS Strong sex effects were identified at both Fmgq2 and Lmgq1, which are also diet-dependent. Interestingly, Fmgq2 and Fmgq3 affect fat gain directly, while Fmgq1 influences fat gain directly and via an intermediate change in serum cholesterol. These results demonstrate how precision nutrition will be advanced through the integration of genetic variation and sex in physiological responses to diets varied in carbohydrate composition.
Collapse
Affiliation(s)
- Anna C. Salvador
- grid.412408.bDepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX USA ,grid.264756.40000 0004 4687 2082Department of Nutrition, Texas A&M University, College Station, TX USA
| | - Danny Arends
- Züchtungsbiologie und molekulare Genetik, Albrecht Daniel Thaer-Institut, Berlin, Germany
| | - William T. Barrington
- grid.412408.bDepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX USA
| | - Ahmed M. Elsaadi
- grid.412408.bDepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX USA
| | - Gudrun A. Brockmann
- Züchtungsbiologie und molekulare Genetik, Albrecht Daniel Thaer-Institut, Berlin, Germany
| | - David W. Threadgill
- grid.412408.bDepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX USA ,grid.264756.40000 0004 4687 2082Department of Nutrition, Texas A&M University, College Station, TX USA ,grid.264756.40000 0004 4687 2082Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX USA
| |
Collapse
|
9
|
Rossi GP, Caroccia B, Seccia TM. Role of estrogen receptors in modulating aldosterone biosynthesis and blood pressure. Steroids 2019; 152:108486. [PMID: 31499072 DOI: 10.1016/j.steroids.2019.108486] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/28/2022]
Abstract
Blood pressure is lower in premenopausal women than in age-matched men; after menopause blood pressure values and the prevalence of hypertension show opposite trends indicating that estrogens contribute to maintaining normal blood pressure values in women. In experimental studies menopause increases aldosterone levels, an effect alleviated by estrogen treatment. We have recently discovered a role of estrogen receptors (ER) in controlling aldosterone biosynthesis in the human adrenocortical zona glomerulosa, which expresses both the classical ERα and β receptors and G protein-coupled estrogen receptor (GPER). We have also identified that GPER mediates an aldosterone-induced aldosterone response. We found that 17 β-estradiol exerts a dual effect: it blunts aldosterone production via ERβ, but displays a potent aldosterone secretagogue effect via GPER activation after ERβ blockade. Thus, in premenopausal women high estrogen levels might tonically blunt aldosterone synthesis via ERβ, thereby maintaining normal blood pressure; after menopause loss of this estrogen-mediated inhibition can contribute to increasing blood pressure via GPER-mediated aldosterone release. The additional findings that GPER mediates an aldosterone-induced stimulation of aldosterone biosynthesis and that GPER predominates in aldosterone-producing adenomas strongly involves this receptor in the pathophysiology of primary aldosteronism. Our purpose here was to provide an update on estrogen receptor function in the normal adrenal cortex and its relevance for the sex differences in blood pressure in light of the newly discovered role of GPER in regulating aldosterone synthesis. The implications of the novel knowledge for the treatment of estrogen-dependent malignancies with ER modulators are also discussed.
Collapse
|
10
|
Moss ME, Carvajal B, Jaffe IZ. The endothelial mineralocorticoid receptor: Contributions to sex differences in cardiovascular disease. Pharmacol Ther 2019; 203:107387. [PMID: 31271793 PMCID: PMC6848769 DOI: 10.1016/j.pharmthera.2019.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease remains the leading cause of death for both men and women. The observation that premenopausal women are protected from cardiovascular disease relative to age-matched men, and that this protection is lost with menopause, has led to extensive study of the role of sex steroid hormones in the pathogenesis of cardiovascular disease. However, the molecular basis for sex differences in cardiovascular disease is still not fully understood, limiting the ability to tailor therapies to male and female patients. Therefore, there is a growing need to investigate molecular pathways outside of traditional sex hormone signaling to fully understand sex differences in cardiovascular disease. Emerging evidence points to the mineralocorticoid receptor (MR), a steroid hormone receptor activated by the adrenal hormone aldosterone, as one such mediator of cardiovascular disease risk, potentially serving as a sex-dependent link between cardiovascular risk factors and disease. Enhanced activation of the MR by aldosterone is associated with increased risk of cardiovascular disease. Emerging evidence implicates the MR specifically within the endothelial cells lining the blood vessels in mediating some of the sex differences observed in cardiovascular pathology. This review summarizes the available clinical and preclinical literature concerning the role of the MR in the pathophysiology of endothelial dysfunction, hypertension, atherosclerosis, and heart failure, with a special emphasis on sex differences in the role of endothelial-specific MR in these pathologies. The available data regarding the molecular mechanisms by which endothelial-specific MR may contribute to sex differences in cardiovascular disease is also summarized. A paradigm emerges from synthesis of the literature in which endothelial-specific MR regulates vascular function in a sex-dependent manner in response to cardiovascular risk factors to contribute to disease. Limitations in this field include the relative paucity of women in clinical trials and, until recently, the nearly exclusive use of male animals in preclinical investigations. Enhanced understanding of the sex-specific roles of endothelial MR could lead to novel mechanistic insights underlying sex differences in cardiovascular disease incidence and outcomes and could identify additional therapeutic targets to effectively treat cardiovascular disease in men and women.
Collapse
Affiliation(s)
- M Elizabeth Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Brigett Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America.
| |
Collapse
|
11
|
Mao B, Wu C, Zheng W, Shen Q, Wang Y, Wang Q, Lin H, Li X, Sun J, Ge RS. Methoxychlor and its metabolite HPTE inhibit rat neurosteroidogenic 3α-hydroxysteroid dehydrogenase and retinol dehydrogenase 2. Neurosci Lett 2018; 684:169-174. [PMID: 30107201 DOI: 10.1016/j.neulet.2018.08.008] [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: 08/05/2017] [Revised: 07/30/2018] [Accepted: 08/09/2018] [Indexed: 01/28/2023]
Abstract
Methoxychlor is primarily used as an insecticide and it is widely present in the environment. The objective of the present study was to investigate the direct effects of methoxychlor and its metabolite hydroxychlor (HPTE) on rat neurosteroidogenic 3α-hydroxysteroid dehydrogenase (AKR1C14) and retinol dehydrogenase 2 (RDH2) activities. Rat AKR1C14 and RDH2 were cloned and expressed in COS-1 cells, and the effects of methoxychlor and HPTE on these enzymes were measured. HPTE was more potent to inhibit AKR1C14 and RDH2 activities than methoxychlor, with IC50 values of 2.602 ± 0.057 μM and 20.473 ± 0.049 μM, respectively, while those of methoxychlor were over 100 μM. HPTE competitively inhibited AKR1C14 and RDH2 when steroid substrates were used, while it showed a mode of mixed inhibition on these enzymes when NADPH/NAD+ were used. We elucidated the binding mode of methoxychlor and HPTE to the crystal structure of AKR1C14 by molecular docking and found that HPTE had higher affinity with the enzyme than methoxychlor. In conclusion, HPTE is more potent than methoxychlor to inhibit both AKR1C14 and RDH2.
Collapse
Affiliation(s)
- Baiping Mao
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chengyun Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China
| | - Wenwen Zheng
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Qiuxia Shen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiyan Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Qiufan Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Han Lin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaoheng Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianliang Sun
- Department of Anesthesia, Hangzhou Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou First People's Hospital, Hangzhou 310006, China.
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
12
|
Haggard DE, Karmaus AL, Martin MT, Judson RS, Woodrow Setzer R, Friedman KP. High-Throughput H295R Steroidogenesis Assay: Utility as an Alternative and a Statistical Approach to Characterize Effects on Steroidogenesis. Toxicol Sci 2018; 162:509-534. [PMID: 29216406 PMCID: PMC10716795 DOI: 10.1093/toxsci/kfx274] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The U.S. Environmental Protection Agency Endocrine Disruptor Screening Program and the Organization for Economic Co-operation and Development (OECD) have used the human adrenocarcinoma (H295R) cell-based assay to predict chemical perturbation of androgen and estrogen production. Recently, a high-throughput H295R (HT-H295R) assay was developed as part of the ToxCast program that includes measurement of 11 hormones, including progestagens, corticosteroids, androgens, and estrogens. To date, 2012 chemicals have been screened at 1 concentration; of these, 656 chemicals have been screened in concentration-response. The objectives of this work were to: (1) develop an integrated analysis of chemical-mediated effects on steroidogenesis in the HT-H295R assay and (2) evaluate whether the HT-H295R assay predicts estrogen and androgen production specifically via comparison with the OECD-validated H295R assay. To support application of HT-H295R assay data to weight-of-evidence and prioritization tasks, a single numeric value based on Mahalanobis distances was computed for 654 chemicals to indicate the magnitude of effects on the synthesis of 11 hormones. The maximum mean Mahalanobis distance (maxmMd) values were high for strong modulators (prochloraz, mifepristone) and lower for moderate modulators (atrazine, molinate). Twenty-five of 28 reference chemicals used for OECD validation were screened in the HT-H295R assay, and produced qualitatively similar results, with accuracies of 0.90/0.75 and 0.81/0.91 for increased/decreased testosterone and estradiol production, respectively. The HT-H295R assay provides robust information regarding estrogen and androgen production, as well as additional hormones. The maxmMd from this integrated analysis may provide a data-driven approach to prioritizing lists of chemicals for putative effects on steroidogenesis.
Collapse
Affiliation(s)
- Derik E. Haggard
- Oak Ridge Institute for Science and Education Postdoctoral Fellow, Oak Ridge, TN. 37831
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| | - Agnes L. Karmaus
- Oak Ridge Institute for Science and Education Postdoctoral Fellow, Oak Ridge, TN. 37831
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| | - Matthew T. Martin
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| | - Richard S. Judson
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| | - R. Woodrow Setzer
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| | - Katie Paul Friedman
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Durham, NC 27711
| |
Collapse
|
13
|
Caroccia B, Seccia TM, Barton M, Rossi GP. Estrogen Signaling in the Adrenal Cortex: Implications for Blood Pressure Sex Differences. Hypertension 2018; 68:840-8. [PMID: 27600178 DOI: 10.1161/hypertensionaha.116.07660] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Brasilina Caroccia
- From the Molecular Internal Medicine, University of Zurich, Switzerland (M.B.); and Department of Medicine-DIMED, University of Padua, Italy (B.C., T.M.S., G.P.R.)
| | - Teresa M Seccia
- From the Molecular Internal Medicine, University of Zurich, Switzerland (M.B.); and Department of Medicine-DIMED, University of Padua, Italy (B.C., T.M.S., G.P.R.)
| | - Matthias Barton
- From the Molecular Internal Medicine, University of Zurich, Switzerland (M.B.); and Department of Medicine-DIMED, University of Padua, Italy (B.C., T.M.S., G.P.R.)
| | - Gian Paolo Rossi
- From the Molecular Internal Medicine, University of Zurich, Switzerland (M.B.); and Department of Medicine-DIMED, University of Padua, Italy (B.C., T.M.S., G.P.R.).
| |
Collapse
|
14
|
Jopek K, Celichowski P, Szyszka M, Tyczewska M, Milecka P, Malendowicz LK, Rucinski M. Transcriptome Profile of Rat Adrenal Evoked by Gonadectomy and Testosterone or Estradiol Replacement. Front Endocrinol (Lausanne) 2017; 8:26. [PMID: 28261157 PMCID: PMC5309227 DOI: 10.3389/fendo.2017.00026] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/30/2017] [Indexed: 01/11/2023] Open
Abstract
Sex differences in adrenal cortex structure and function are well known in different species. In the rat, they are manifested as larger adrenal cortex and higher corticosterone secretion by females compared with males. These sex differences depend, among others, on functioning of the hypothalamic-pituitary-adrenal axis (HPA). In this aspect, it is widely accepted that testosterone exerts an inhibitory and estradiol stimulatory effect on the said axis. The molecular bases of these sex-related differences are poorly understood. Therefore, we performed studies aimed to demonstrate the effect of testosterone and estradiol on the expression of differentially regulated genes in rat adrenal gland. The classical method applied in the study-gonadectomy and gonadal hormone replacement-allows obtaining results suggesting a physiological role of the tested hormone (testosterone or estradiol) in the regulation of the specific genes. Adult male and female rats were either gonadectomized or sham operated. Half of orchiectomized rats were replaced with testosterone while ovariectomized ones with estradiol. Transcriptome was identified by means of Affymetrix® Rat Gene 2.1 ST Array. Differentially expressed genes were analyzed by means of DAVID web-based bioinformatic tools and confirmed by means of Gene Set Enrichment Analysis. For selected genes, validation of the results was performed using QPCR. Performed experiments have provided unexpected results. Contrary to expectations, in orchiectomized rats, testosterone replacement stimulates expression of numerous genes, mainly those associated with lipids and cholesterol metabolism. However, in ovariectomized animals, estradiol replacement inhibits the expression of genes, mainly those involved in intracellular signaling pathways. The physiological relevance of these findings awaits further research.
Collapse
Affiliation(s)
- Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| | - Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| | - Paulina Milecka
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| | - Ludwik K. Malendowicz
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
- *Correspondence: Ludwik K. Malendowicz,
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Science, Poznan, Poland
| |
Collapse
|
15
|
Caroccia B, Seccia TM, Campos AG, Gioco F, Kuppusamy M, Ceolotto G, Guerzoni E, Simonato F, Mareso S, Lenzini L, Fassina A, Rossi GP. GPER-1 and estrogen receptor-β ligands modulate aldosterone synthesis. Endocrinology 2014; 155:4296-304. [PMID: 25167221 DOI: 10.1210/en.2014-1416] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fertile women have lower blood pressure and cardiovascular risk than age-matched men, which suggests that estrogens exert cardiovascular protective effects. However, whether 17 β-estradiol (E2) blunts aldosterone secretion, and thereby affects the gender dimorphism of blood pressure, is unknown. We therefore sought for the estrogen receptor (ER) subtypes in human adrenocortical tissues ex vivo by performing gene and protein expression studies. We also investigated the effect of E2 on aldosterone synthesis and the involved receptors through in vitro functional experiments in the adrenocortical cells HAC15. We found that in the human adrenal cortex and aldosterone-producing adenoma cells, the most expressed ERs were the ERβ and the G protein-coupled receptor-1 (GPER-1), respectively. After selective ERβ blockade, E2 (10 nmol/L) markedly increased both the expression of aldosterone synthase and the production of aldosterone (+5- to 7-fold vs baseline, P < .001). Under the same condition, the GPER-1 receptor agonist 1-[4-(6-bromo-benzo (1, 3)dioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c] quinolin-8-yl]-ethanone (G-1) (10 nmol/L) mimicked this effect, which was abrogated by cotreatment with either the GPER-1 receptor antagonist (3aS*,4R*,9bR*)-4-(6-Bro-mo-1,3-benzodioxol-5-yl)-3a,4,5,9b-3H-cyclopenta[c]quinoline (G-15), or a selective protein kinase A inhibitor 8-Bromo-2-monobutyryladenosine-3,5-cyclic mono-phosphorothioate, Rp-isomer. Silencing of the ERβ significantly raised aldosterone synthase expression and aldosterone production. Conversely, silencing of the GPER-1 lowered aldosterone synthase gene and protein expression. Moreover, it blunted the stimulatory effect of E2 on aldosterone synthase that was seen during ERβ blockade. These results support the conclusion that in humans, E2 inhibits aldosterone synthesis by acting via ERβ. Pharmacologic disinhibition of ERβ unmasks a potent secretagogue effect of E2 that involves GPER-1 and protein kinase A signaling.
Collapse
Affiliation(s)
- Brasilina Caroccia
- Internal Medicine 4 (B.C., T.M.S., A.G.C., F.G., M.K., G.C., E.G., S.M., L.L., G.P.R.), Department of Medicine-DIMED, University of Padua, Padua 35126, Italy; and Surgical Pathology and Cytopathology Unit (F.S., A.F.), Department of Medicine, University of Padua, Padua 35126, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Pospíšilová H, Vaňková M, Hill M, Meloun M, Bendlová B, Dušková M, Stárka L. The differences between aromatizable and non-aromatizable androgens in relation to body composition and metabolic syndrome risk factors in men. J Steroid Biochem Mol Biol 2012; 132:105-11. [PMID: 22421634 DOI: 10.1016/j.jsbmb.2012.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/21/2012] [Accepted: 02/26/2012] [Indexed: 11/30/2022]
Abstract
The relationships between the parameters of metabolic syndrome and non-aromatizable metabolites of testosterone have been discussed in literature. Some papers describe these metabolites as one of the possible causes of male-type obesity. On the contrary, other studies show a protective influence of dihydrotestosterone on visceral obesity. The aim of this study to analyse the relationship between anthropometric parameters, lipid spectrum, glycemia and the level of endogenous testosterone and dihydrotestosterone, and to compare the effects of these androgens. Our population-based study involved 232 healthy men ranging from 20 to 78 years with BMI 18 to 39 kg/m(2). Serum testosterone, dihydrotestosterone and sex hormone binding globulin SHBG levels, lipid spectrum, glucose metabolism parameters were measured and the oral glucose tolerance test was carried out in all subjects. Their anthropometric parameters (weight, height, waist, hips, waist-to-hip ratio, 14 skin folds) and body composition parameters were determined and calculated by the Antropo program. Multiple regression analysis showed a correlation between hormonal levels, esp. of testosterone and dihydrotestosterone, and the anthropometric data, lipid spectrum and parameters of glucose regulation. Low testosterone and/or dihydrotestosterone was correlated to a higher body-mass index, fat content, waist diameter, total-, HDL-, LDL-cholesterol and triglycerides, fasting glucose, insulin resistance and lower muscle and bone mass. In addition, statistical analysis using multivariate regression with reduction in dimensionality did not discover any striking difference between aromatizable and non-aromatizable androgens in their association to lipid and glucose metabolism parameters in healthy, normosthenic men. In conclusion, the association of endogenous testosterone and dihydrotestosterone to anthropometric data, lipid spectrum and insulin sensitivity are of the same quality; however, the effect of the circulating levels of dihydrotestosterone is quantitatively smaller.
Collapse
|
17
|
Effects of CYP7B1-related steroids on androgen receptor activation in different cell lines. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:973-9. [PMID: 22484622 DOI: 10.1016/j.bbalip.2012.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 02/29/2012] [Accepted: 03/05/2012] [Indexed: 11/24/2022]
Abstract
The widely expressed steroid hydroxylase CYP7B1 is involved in metabolism of a number of steroids reported to influence estrogen and androgen signaling. Several studies by us and other investigators have linked this enzyme to effects on estrogen receptor activation. In a previous report we examined the effect of CYP7B1-mediated hormone metabolism for estrogen-mediated response in kidney-derived HEK293 cells. In the current study we used an androgen response element (ARE) reporter system to examine androgen-dependent response of some CYP7B1 substrates and CYP7B1-formed metabolites in several cell lines derived from different tissues. The results indicate significantly lower androgen receptor activation by CYP7B1-formed steroid metabolites than by the corresponding steroid substrates, suggesting that CYP7B1-mediated catalysis may decrease some androgenic responses. Thus, CYP7B1-dependent metabolism may be of importance not only for estrogenic signaling but also for androgenic. This finding, that CYP7B1 activity may be a regulator of androgenic signaling by converting AR ligands into less active metabolites, is also supported by real-time RT-PCR experiment where a CYP7B1 substrate, but not the corresponding product, was able to stimulate known androgen-sensitive genes. Furthermore, our data indicate that the effects of some steroids on hormone response element reporter systems are cell line-specific. For instance, despite transfection of the same reporter systems, 5-androstene-3β,17β-diol strongly activates an androgen-dependent response element in prostate cancer cells whereas it elicits only ER-dependent responses in kidney HEK293 cells. Potential roles of cell-specific metabolism or comodulator expression for the observed differences are discussed.
Collapse
|
18
|
DUŠKOVÁ M, POSPÍŠILOVÁ H. The Role of Non-Aromatizable Testosterone Metabolite in Metabolic Pathways. Physiol Res 2011; 60:253-61. [DOI: 10.33549/physiolres.932080] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Dihydrotestosterone (DHT) originates via irreversible reduction of testosterone by catalytic activity of 5α-reductase enzyme and it is demonstratively the most effective androgen. Androgens influence adipose tissue in men either directly by stimulation of the androgen receptor or indirectly, after aromatization, by acting at the estrogen receptor. DHT as a non-aromatizable androgen could be responsible for a male type fat distribution. The theory of non-aromatizable androgens as a potential cause of a male type obesity development has been studied intensively. However, physiological levels of DHT inhibit growth of mature adipocytes. In animal models, substitution of DHT in males after gonadectomy has a positive effect on body composition as a testosterone therapy. Thus, DHT within physiological range positively influences body composition. However, there are pathological conditions with an abundance of DHT, e.g. androgenic alopecia and benign prostatic hyperplasia. These diseases are considered as risk factors for development of metabolic syndrome or atherosclerosis. In obese people, DHT metabolism in adipose tissue is altered. Local abundance of non-aromatizable androgen has a negative effect on adipose tissue and it could be involved in pathogenesis of metabolic and cardiovascular diseases. Increased DHT levels, compared to physiological levels, have negative effect on development of cardiovascular diseases. Difference between the effect of physiological and increased level brings about certain paradox.
Collapse
Affiliation(s)
- M. DUŠKOVÁ
- Institute of Endocrinology, Prague, Czech Republic
| | | |
Collapse
|
19
|
Current Opinion in Endocrinology, Diabetes & Obesity. Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:293-312. [PMID: 20418721 DOI: 10.1097/med.0b013e328339f31e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|