1
|
Peck M, Connelly P, Lucas-Herald AK. Cardiometabolic outcomes of early onset hypogonadism in males. Best Pract Res Clin Endocrinol Metab 2025:102004. [PMID: 40399185 DOI: 10.1016/j.beem.2025.102004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Testosterone is an important vascular hormone, with multiple effects reported on the vasculature. As such, boys and men with early onset hypogonadism may have altered cardiovascular function, with the potential to result in adverse cardiometabolic outcomes in adulthood. Given the fact that cardiovascular changes in the young can affect future cardiovascular health, there is a need to better understand the influence of androgens on the vasculature in those with conditions such as 46, XY Disorders of Sex Development and Klinefelter Syndrome. This review summarises what is known about hypogonadism and the effects of testosterone supplementation in adults with hypogonadism, as well as what is currently understood in those with early onset hypogonadism specifically. A number of research gaps persist in this area and there is a need for international collaborative studies to address these for future generations of affected individuals.
Collapse
Affiliation(s)
- Mariska Peck
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, 1345 Govan Road, Glasgow G51 4TF, UK.
| | - Paul Connelly
- Department of Endocrinology, Queen Elizabeth University Hospital, Govan Road, 1345 Govan Road, Glasgow G51 4TF, UK.
| | - Angela K Lucas-Herald
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, 1345 Govan Road, Glasgow G51 4TF, UK.
| |
Collapse
|
2
|
Adams ZH, Berbrier DE, Schwende BK, Huckins W, Richards CT, Rees DA, Usselman CW, Lord RN. The impact of androgens on cardiovascular control mechanisms in polycystic ovary syndrome: Recent advances and translational approaches. J Physiol 2025; 603:2937-2957. [PMID: 40321041 DOI: 10.1113/jp287288] [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: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 06/02/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy in premenopausal females. The condition is associated with an increased prevalence of cardiovascular risk factors, including hypertension. Observational studies report that some blood pressure control mechanisms are altered in PCOS compared to controls (sympathetic nervous system activity, endothelial and vasodilator function, renin angiotensin aldosterone system activation), and that these impairments correlate with androgen hormone levels, which are chronically elevated in the condition. As such, hyperandrogenism is the proposed locus of origin for the link between PCOS and cardiovascular dysfunction, yet the underlying mechanisms remain poorly understood. Preclinical work has provided some insight into how androgens modulate blood pressure control in PCOS. However there are marked discrepancies between the effects of androgens in cellular and tissue studies versus in vivo animal and human PCOS studies. This may be related to the heterogeneity of the preclinical models and samples used in this research and whether preclinical work is modelling hyperandrogenism in physiologically relevant terms for PCOS. This review collates preclinical and clinical evidence to summarise what is known and what remains unknown about cardiovascular control mechanisms in PCOS. We examine aspects of blood pressure regulation that are altered in other hypertensive cohorts, presenting current evidence for a mechanistic role of androgens on these systems, while acknowledging the diverse experimental models and participant cohorts from which the results are derived.
Collapse
Affiliation(s)
- Zoe H Adams
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Danielle E Berbrier
- Department of Kinesiology and Physical Education, McGill University, Montreal, Canada
| | - Brittany K Schwende
- Department of Kinesiology and Physical Education, McGill University, Montreal, Canada
| | - Will Huckins
- Department of Kinesiology and Physical Education, McGill University, Montreal, Canada
| | - Cory T Richards
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - D Aled Rees
- Cardiff and Vale University Health Board, Cardiff, UK
- Neuroscience and Mental Health Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Charlotte W Usselman
- Department of Kinesiology and Physical Education, McGill University, Montreal, Canada
| | - Rachel N Lord
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
3
|
Hornig N, Batista RL. Androgen insensitivity and the evolving genetic heterogeneity. Best Pract Res Clin Endocrinol Metab 2025:102000. [PMID: 40335402 DOI: 10.1016/j.beem.2025.102000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Androgen Insensitivity Syndrome (AIS) is a 46,XY difference of sex development (DSD) classically caused by mutations in the androgen receptor (AR) gene, leading to variable androgen resistance and a broad phenotypic spectrum traditionally classified as complete, partial, or mild. Phenotypic variability can occur even with identical AR mutations, particularly those within the ligand-binding domain of the AR. Emerging evidence implicates non-coding regulatory variants, deep intronic mutations, AR co-regulator dysfunction, and oligogenic inheritance in the aetiology of AIS. The molecular diagnostic workflow should incorporate either targeted AR sequencing or whole-exome sequencing, depending on the clinical context. Biochemical and functional assays remain clinically useful, especially when AR variants are not detected or when variants of unknown significance (VUS) are identified. Advances in patient-derived hiPSC models and testicular organoids provide new insights into AR function and therapeutic strategies. Expanding genomic and epigenetic research will refine diagnostic accuracy, and personalized care, ultimately optimizing patient outcomes in AIS.
Collapse
Affiliation(s)
- Nadine Hornig
- Institute of Human Genetics, Christian Albrechts University of Kiel (CAU) and University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Rafael Loch Batista
- Developmental Endocrinology Unit and Laboratório de Investigações Médicas (LIM/42), Endocrinology Division, Internal Medicine Department, Medical School, University of São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
4
|
Chevalley T, Dübi M, Fumeaux L, Merli MS, Sarre A, Schaer N, Simeoni U, Yzydorczyk C. Sexual Dimorphism in Cardiometabolic Diseases: From Development to Senescence and Therapeutic Approaches. Cells 2025; 14:467. [PMID: 40136716 PMCID: PMC11941476 DOI: 10.3390/cells14060467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
The global incidence and prevalence of cardiometabolic disorders have risen significantly in recent years. Although lifestyle choices in adulthood play a crucial role in the development of these conditions, it is well established that events occurring early in life can have an important effect. Recent research on cardiometabolic diseases has highlighted the influence of sexual dimorphism on risk factors, underlying mechanisms, and response to therapies. In this narrative review, we summarize the current understanding of sexual dimorphism in cardiovascular and metabolic diseases in the general population and within the framework of the Developmental Origins of Health and Disease (DOHaD) concept. We explore key risk factors and mechanisms, including the influence of genetic and epigenetic factors, placental and embryonic development, maternal nutrition, sex hormones, energy metabolism, microbiota, oxidative stress, cell death, inflammation, endothelial dysfunction, circadian rhythm, and lifestyle factors. Finally, we discuss some of the main therapeutic approaches, responses to which may be influenced by sexual dimorphism, such as antihypertensive and cardiovascular treatments, oxidative stress management, nutrition, cell therapies, and hormone replacement therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine Yzydorczyk
- Developmental Origins of Health and Disease (DOHaD) Laboratory, Division of Pediatrics, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (T.C.); (M.D.); (L.F.); (M.S.M.); (A.S.); (N.S.)
| |
Collapse
|
5
|
Krysiak R, Kowalcze K, Szkróbka W, Okopień B. Vitamin D Status Determines Cardiometabolic Effects of Testosterone Replacement Therapy in Men with Late-Onset Hypogonadism. Nutrients 2025; 17:1013. [PMID: 40290009 PMCID: PMC11944846 DOI: 10.3390/nu17061013] [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: 02/11/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Low testosterone levels and low vitamin D status are associated with increased cardiometabolic risk. The purpose of this study was to investigate whether vitamin D status determines the cardiometabolic effects of testosterone replacement therapy. Methods: The study population consisted of three groups of men with late-onset hypogonadism: vitamin D-naive individuals with 25-hydroxyvitamin D levels between 20 and 30 ng/mL (group I), males with 25-hydroxyvitamin D levels between 30 and 60 ng/mL receiving vitamin D supplementation because of previous low vitamin D status (group II), and vitamin D-naïve subjects with 25-hydroxyvitamin D levels between 30 and 60 ng/mL (group III). Circulating levels of total testosterone, 25-hydroxyvitamin D, glucose, insulin, lipids, uric acid, high-sensitivity C-reactive protein (hsCRP), homocysteine, fibrinogen, and urinary albumin-to-creatinine ratio (UACR) were assessed before and six months after intramuscular testosterone administration (250 mg every three weeks). Results: Group I differed from the remaining groups in baseline values of 25-hydroxyvitamin D, hsCRP, homocysteine, fibrinogen, UACR, and the Framingham Risk Score. In all three groups, testosterone injections increased plasma testosterone levels and had a neutral effect on 25-hydroxyvitamin D concentration. In groups II and III, the drug improved insulin sensitivity and reduced LDL cholesterol, uric acid, hsCRP, homocysteine, fibrinogen, and UACR. In group I, the impact of testosterone was limited to a small decrease in HDL cholesterol and hsCRP. Only in groups II and III did testosterone reduce the Framingham Risk Score. There were no differences in the strength of testosterone action between both groups. In groups II and III, the replacement-induced changes in insulin sensitivity, LDL cholesterol, uric acid, hsCRP, homocysteine, fibrinogen, UACR, and the Framingham Risk Score positively correlated with 25-hydroxyvitamin D concentration. Conclusions: The study results suggest that the cardiometabolic effects of exogenous testosterone in men with testosterone deficiency may be determined by vitamin D status.
Collapse
Affiliation(s)
- Robert Krysiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (W.S.); (B.O.)
| | - Karolina Kowalcze
- Department of Pediatrics in Bytom, Faculty of Health Sciences in Katowice, Medical University of Silesia, Stefana Batorego 15, 41-902 Bytom, Poland;
- Department of Pathophysiology, Faculty of Medicine, Academy of Silesia, Rolna 43, 40-555 Katowice, Poland
| | - Witold Szkróbka
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (W.S.); (B.O.)
| | - Bogusław Okopień
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (W.S.); (B.O.)
| |
Collapse
|
6
|
De Silva NL, Papanikolaou N, Grossmann M, Antonio L, Quinton R, Anawalt BD, Jayasena CN. Male hypogonadism: pathogenesis, diagnosis, and management. Lancet Diabetes Endocrinol 2024; 12:761-774. [PMID: 39159641 DOI: 10.1016/s2213-8587(24)00199-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 08/21/2024]
Abstract
Organic male hypogonadism due to irreversible hypothalamic-pituitary-testicular (HPT) pathology is easily diagnosed and treated with testosterone-replacement therapy. However, controversy surrounds the global practice of prescribing testosterone to symptomatic men with low testosterone and non-gonadal factors reducing health status, such as obesity, type 2 diabetes, and ageing (ie, functional hypogonadism), but without identifiable HPT axis pathology. Health optimisation remains the gold-standard management strategy. Nevertheless, in the last decade large clinical trials and an individual patient data meta-analysis of smaller clinical trials confirmed that testosterone therapy induces modest, yet statistically significant, improvements in sexual function without increasing short-term to medium-term cardiovascular or prostate cancer risks in men with functional hypogonadism. Although testosterone improves bone mineral density and insulin sensitivity in these men, trials from the last decade suggest insufficient evidence to determine the safety and effectiveness of use of this hormone for the prevention of fractures or type 2 diabetes. This Review discusses the pathogenesis and diagnosis of male hypogonadism and appraises the evidence underpinning the management of this condition.
Collapse
Affiliation(s)
- Nipun Lakshitha De Silva
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Faculty of Medicine, General Sir John Kotelawala Defence University, Colombo, Sri Lanka
| | - Nikoleta Papanikolaou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Mathis Grossmann
- Department of Medicine (Austin Health), The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology, Austin Health, Heidelberg, VIC, Australia
| | - Leen Antonio
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium; Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Richard Quinton
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Northern Regional Gender Dysphoria Service, Cumbria Northumberland Tyne & Wear NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Bradley David Anawalt
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Channa N Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
7
|
de Silva NL, Grant B, Minhas S, Jayasena CN. Cardiovascular disease and testosterone therapy in male hypogonadism. Ann N Y Acad Sci 2024; 1540:121-132. [PMID: 39243393 DOI: 10.1111/nyas.15211] [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] [Indexed: 09/09/2024]
Abstract
This review assesses the evidence of the physiological effects of testosterone on cardiovascular health, the association between male hypogonadism and cardiovascular health, and the effects of testosterone therapy on cardiovascular health in male hypogonadism. Preclinical studies suggest complex effects of testosterone on cardiovascular risk by acting on skeletal muscle, cardiomyocytes, vasculature, adipocytes, insulin action, and erythropoiesis. Furthermore, low testosterone has a bi-directional association with cardiometabolic risk. Observational studies have reported worse metabolic profiles in men with organic hypogonadism. However, a consistent association between major cardiovascular events and male hypogonadism has not been established. Hematocrit increases with testosterone therapy; however, most studies do not report an increase in venous thromboembolism risk. Although some observational studies and a small randomized controlled study reported an increased risk of cardiovascular disease, recent data confirm the medium-term cardiovascular safety of testosterone therapy in middle-aged and older men with low testosterone.
Collapse
Affiliation(s)
- Nipun Lakshitha de Silva
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Faculty of Medicine, General Sir John Kotelawala Defence University, Colombo, Sri Lanka
| | - Bonnie Grant
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Suks Minhas
- Department of Urology, Imperial College Healthcare NHS Trust, London, UK
| | - Channa N Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
8
|
Dinakaran A, AR S, Rajagambeeram R, Nanda SK, Daniel M. SHBG and Insulin resistance - Nexus revisited. Bioinformation 2024; 20:816-821. [PMID: 39411775 PMCID: PMC11471403 DOI: 10.6026/973206300200816] [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: 08/01/2024] [Revised: 08/31/2024] [Accepted: 08/31/2024] [Indexed: 10/19/2024] Open
Abstract
Sex hormone binding globulin (SHBG) is a liver-synthesized glycoprotein. Low SHBG levels are associated with insulin resistance (IR). Specific single nucleotide polymorphisms (SNPs) in the SHBG gene are linked to IR. Therefore, it is of interest to provide a review on the comprehensive overview for SHBG related to IR.
Collapse
Affiliation(s)
- Asha Dinakaran
- Department of Biochemistry, Pondicherry Institute of Medical Sciences, Kalapet, Pondicherry, India
| | - Srinivasan AR
- Department of Biochemistry, Mahatma Gandhi Medical College & Research Institute, Sri Balaji Vidyapeeth, Deemed to be University, Pillaiyarkuppam, Pondicherry, India
| | - Reeta Rajagambeeram
- Department of Biochemistry, Mahatma Gandhi Medical College & Research Institute, Sri Balaji Vidyapeeth, Deemed to be University, Pillaiyarkuppam, Pondicherry, India
| | - Sunil Kumar Nanda
- Department of Biochemistry, Pondicherry Institute of Medical Sciences, Kalapet, Pondicherry, India
| | - Mary Daniel
- Department of Obstetrics & Gynaecology, Pondicherry Institute of Medical Sciences, Kalapet, Pondicherry, India
| |
Collapse
|
9
|
Barjesteh F, Heidari-Kalvani N, Alipourfard I, Najafi M, Bahreini E. Testosterone, β-estradiol, and hepatocellular carcinoma: stimulation or inhibition? A comparative effect analysis on cell cycle, apoptosis, and Wnt signaling of HepG2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6121-6133. [PMID: 38421409 DOI: 10.1007/s00210-024-03019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
Unlike breast and prostate cancers, which are specifically affected by estrogens or androgens, hepatocellular carcinoma has been reported to be influenced by both sex hormones. Given the coincidental differences of hepatocellular carcinoma in men and women, we investigated the effects of β-estradiol and testosterone on the cell cycle, apoptosis, and Wnt signaling in a model of hepatocellular carcinoma to understand the sex hormone-related etiology. To determine the effective concentration of both hormones, an MTT assay was performed. The effects of β-estradiol and testosterone on cell proliferation and death were evaluated by specific staining and flow cytometry. In addition, gene expression levels of estimated factors involved in GPC3-Wnt survival signaling were analyzed using quantitative real-time polymerase chain reaction. Both hormones inhibited hepatic cell proliferation through arresting the cell cycle at S/G2 and increased the apoptosis rate in HepG2 cells. Both hormones dose-dependently decreased GPC3, Wnt, and DVL expression levels as activators of the Wnt-signaling pathway. In the case of Wnt-signaling inhibitors, the effects of both hormones on WIF were negligible, but they increased DKK1 levels in a dose-dependent manner. In each of the effects mentioned above, β-estradiol was notably more potent than testosterone. In contrast to the primary hypothesis of the project, in which testosterone was considered a stimulating carcinogenic factor in HCC pathogenesis, testosterone inhibited the occurrence of HCC similarly to β-estradiol. However, this inhibitory effect was weaker than that of β-estradiol and requires further study.
Collapse
Affiliation(s)
- Fereshteh Barjesteh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Nafiseh Heidari-Kalvani
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Mohammad Najafi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran.
| |
Collapse
|
10
|
Xega V, Liu JL. Beyond reproduction: unraveling the impact of sex hormones on cardiometabolic health. MEDICAL REVIEW (2021) 2024; 4:284-300. [PMID: 39135604 PMCID: PMC11317208 DOI: 10.1515/mr-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/07/2024] [Indexed: 08/15/2024]
Abstract
This review thoroughly explores the multifaceted roles of sexual hormones, emphasizing their impact beyond reproductive functions and underscoring their significant influence on cardiometabolic regulation. It analyzes the broader physiological implications of estrogen, testosterone, and progesterone, highlighting their effects on metabolic syndrome, lipid metabolism, glucose homeostasis, and cardiovascular health. Drawing from diverse molecular, clinical, and therapeutic studies, the paper delves into the intricate interplay between these hormones and cardiometabolic processes. By presenting a comprehensive analysis that goes beyond traditional perspectives, and recognizing sexual hormones as more than reproductive agents, the review sheds light on their broader significance in health and disease management, advocating for holistic and personalized medical approaches.
Collapse
Affiliation(s)
- Viktoria Xega
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Jun-Li Liu
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
11
|
Mu X, Liu S, Wang Z, Jiang K, McClintock T, Stromberg AJ, Tezanos AV, Lee ES, Curci JA, Gong MC, Guo Z. Androgen aggravates aortic aneurysms via suppression of PD-1 in mice. J Clin Invest 2024; 134:e169085. [PMID: 38900572 PMCID: PMC11290977 DOI: 10.1172/jci169085] [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: 01/23/2023] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Androgen has long been recognized for its pivotal role in the sexual dimorphism of cardiovascular diseases, including aortic aneurysms (AAs), a devastating vascular disease with a higher prevalence and fatality rate in men than in women. However, the mechanism by which androgen mediates AAs is largely unknown. Here, we found that male, not female, mice developed AAs when exposed to aldosterone and high salt (Aldo-salt). We revealed that androgen and androgen receptors (ARs) were crucial for this sexually dimorphic response to Aldo-salt. We identified programmed cell death protein 1 (PD-1), an immune checkpoint, as a key link between androgen and AAs. Furthermore, we demonstrated that administration of anti-PD-1 Ab and adoptive PD-1-deficient T cell transfer reinstated Aldo-salt-induced AAs in orchiectomized mice and that genetic deletion of PD-1 exacerbated AAs induced by a high-fat diet and angiotensin II (Ang II) in nonorchiectomized mice. Mechanistically, we discovered that the AR bound to the PD-1 promoter to suppress the expression of PD-1 in the spleen. Thus, our study unveils a mechanism by which androgen aggravates AAs by suppressing PD-1 expression in T cells. Moreover, our study suggests that some patients with cancer might benefit from screenings for AAs during immune checkpoint therapy.
Collapse
Affiliation(s)
- Xufang Mu
- Departments of Pharmacology and Nutritional Sciences
| | | | - Zhuoran Wang
- Departments of Pharmacology and Nutritional Sciences
| | | | | | | | | | - Eugene S. Lee
- Department of Research, Sacramento Veterans Affairs Medical Center, Mather, California, USA
| | - John A. Curci
- Department of Vascular Surgery, Vanderbilt University, Nashville, Tennessee, USA
| | - Ming C. Gong
- Physiology, and
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Zhenheng Guo
- Departments of Pharmacology and Nutritional Sciences
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Research, Lexington Veterans Affairs Medical Center, Lexington, Kentucky, USA
| |
Collapse
|
12
|
Carbajal-García A, Reyes-García J, Díaz-Hernández V, Casas-Hernández MF, Flores-Murrieta FJ, Montaño LM. Testosterone Enhances K V Currents and Airway Smooth Muscle Relaxation Induced by ATP and UTP through P2Y 4 Receptors and Adenylyl Cyclase Pathway. Int J Mol Sci 2024; 25:4652. [PMID: 38731872 PMCID: PMC11083821 DOI: 10.3390/ijms25094652] [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: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
Numerous studies suggest the involvement of adenosine-5'-triphosphate (ATP) and similar nucleotides in the pathophysiology of asthma. Androgens, such as testosterone (TES), are proposed to alleviate asthma symptoms in young men. ATP and uridine-5'-triphosphate (UTP) relax the airway smooth muscle (ASM) via purinergic P2Y2 and P2Y4 receptors and K+ channel opening. We previously demonstrated that TES increased the expression of voltage-dependent K+ (KV) channels in ASM. This study investigates how TES may potentiate ASM relaxation induced by ATP and UTP. Tracheal tissues treated with or without TES (control group) from young male guinea pigs were used. In organ baths, tracheas exposed to TES (40 nM for 48 h) showed enhanced ATP- and UTP-evoked relaxation. Tetraethylammonium, a K+ channel blocker, annulled this effect. Patch-clamp experiments in tracheal myocytes showed that TES also increased ATP- and UTP-induced K+ currents, and this effect was abolished with flutamide (an androgen receptor antagonist). KV channels were involved in this phenomenon, which was demonstrated by inhibition with 4-aminopyridine. RB2 (an antagonist of almost all P2Y receptors except for P2Y2), as well as N-ethylmaleimide and SQ 22,536 (inhibitors of G proteins and adenylyl cyclase, respectively), attenuated the enhancement of the K+ currents induced by TES. Immunofluorescence and immunohistochemistry studies revealed that TES did not modify the expression of P2Y4 receptors or COX-1 and COX-2, while we have demonstrated that this androgen augmented the expression of KV1.2 and KV1.5 channels in ASM. Thus, TES leads to the upregulation of P2Y4 signaling and KV channels in guinea pig ASM, enhancing ATP and UTP relaxation responses, which likely limits the severity of bronchospasm in young males.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Verónica Díaz-Hernández
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - María F. Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Francisco Javier Flores-Murrieta
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| |
Collapse
|
13
|
Shah I, Hakeem MK, Alraeesi A, Barker J. Innovative Detection of Testosterone Esters in Camel Hair: Unravelling the Mysteries of Dromedary Endocrinology. Molecules 2023; 29:97. [PMID: 38202682 PMCID: PMC10779721 DOI: 10.3390/molecules29010097] [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: 10/05/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Introduction: Doping and steroid use represent a serious threat to animal health and can even lead to their untimely and painful death. However, doping is an acute problem in today's animal racing world, particularly in camel racing. Testosterone and its ten esters (benzoate, valerate, isocaproate, hexahydrobenzoate, decanoate, undecanoate, laurate, enanthate, cypionate, and caproate) are of utmost importance, because when they are administered to animals it is difficult to measure them efficiently. The levels of testosterone and its esters in camels and other animals are typically determined using urine and blood tests. The aim of this study was to develop and validate a liquid chromatographic-mass spectrometric (LC-MS/MS) method to determine testosterone esters in camel hair, and to apply the validated method to determine testosterone esters in collected samples. To our knowledge, this is the first report of such research. Results and Discussion: The levels of testosterone and its ten derivatives, along with the cortisol-D4 internal standard, were optimised for LC-MS/MS analysis; however, only testosterone along with its seven esters (namely benzoate, valerate, isocaproate, hexahydrobenzoate, decanoate, undecanoate and laurate) could be validated in camel hair. Only five testosterone esters could be determined in camel hair samples; the concentrations were obtained as 10.5-14.9 pg/mg for valerate (in three camels), 12.5-151.6 pg/mg for hexahydrobenzoate (in six camels), 4.8-32.1 pg/mg for laurate (in five camels), 5.1 pg/mg decanoate (in one camel), and 8.35-169 pg/mg for testosterone (in all 24 camels). Interestingly, the three racing camels displayed high concentrations of testosterone (59.2-169 pg/mg, all three camels), laurate (4.8-14.5 pg/mg, two camels), hexahydrobenzoate (116 pg/mg, one camel), decanoate (5.1 pg/mg, one camel), and valerate (11.7 pg/mg, one camel). Methods: Camel hair samples were collected from 21 non-racing dromedary camels along with three racing camels in Al Ain, UAE; these were decontaminated, pulverised, sonicated, and extracted prior to analysis. An LC-MS/MS method was employed to determine the levels of testosterone esters in the hair samples. Conclusions: This novel camel-hair test procedure is accurate, sensitive, rapid, and robust. The findings reported in this study could be significant to evaluate racing camels for suspected doping offenses. Further controlled testosterone supplementation studies are required to evaluate individual esters' effects on camel health and diseases and on performance enhancement levels. This new hair test could promote further studies in doping control, toxicology, and pharmacology, as well as having other clinical applications relating to camel health, injury, and disease.
Collapse
Affiliation(s)
- Iltaf Shah
- Department of Chemistry, College of Science, UAE University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.H.); (A.A.)
| | - Muhammad K. Hakeem
- Department of Chemistry, College of Science, UAE University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.H.); (A.A.)
| | - Aysha Alraeesi
- Department of Chemistry, College of Science, UAE University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.H.); (A.A.)
| | - James Barker
- School of Pharmacy and Chemistry, Kingston University, Kingston upon Thames KT1 2EE, UK;
| |
Collapse
|
14
|
Cheng L, Wang S. Lower serum testosterone is associated with increased likelihood of arthritis. Sci Rep 2023; 13:19241. [PMID: 37935765 PMCID: PMC10630339 DOI: 10.1038/s41598-023-46424-1] [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: 05/12/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023] Open
Abstract
Studies have suggested that serum testosterone levels may be strongly correlated with the pathogenesis of arthritis. Therefore, the aim of this study was to assess the relationship between serum testosterone levels and arthritis in US adults using the National Health and Nutrition Examination Survey (NHANES). We used the database from NHANES, 2013-2016 to perform a cross-sectional study. This study investigated the relationship between serum testosterone and arthritis using multivariate logistic regression models and also used smoothed curve fitting and generalized additivity models. A total of 10,439 adults were included in this analysis. A significant negative association between serum testosterone and arthritis was found in a linear regression analysis. The study showed that the arthritis group had lower testosterone levels than the non-arthritis group. The univariate multivariate analyses of Q4, using Q1 as a reference, all showed a significantly lower risk of developing arthritis. In subgroup analyses, the negative correlation between serum testosterone levels and arthritis was more significant in women and those with a body mass index (BMI) ≥ 30 kg/m2. After controlling for various variables, we found a significant association between serum testosterone and arthritis in this analysis. Further study of the relationship between testosterone and arthritis is necessary to clarify the specific mechanism of serum testosterone action on arthritis.
Collapse
Affiliation(s)
- Lulu Cheng
- College of Acupuncture-Moxibustion and Tuina, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Graduate School, Wuhan Sports University, Wuhan, 430079, China.
| | - Siyu Wang
- Graduate School, Wuhan Sports University, Wuhan, 430079, China
| |
Collapse
|
15
|
Murphy CN, Delles C, Davies E, Connelly PJ. Cardiovascular disease in transgender individuals. Atherosclerosis 2023; 384:117282. [PMID: 37821271 DOI: 10.1016/j.atherosclerosis.2023.117282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/23/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023]
Abstract
The population of people identifying as transgender has grown rapidly in recent years, resulting in a substantive increase in individuals obtaining gender-affirming medical care to align their secondary sex characteristics with their gender identity. This has established benefits for patients including improvements in gender dysphoria and psychosocial functioning, while reducing adverse mental health outcomes. Despite these potential advantages, recent evidence has suggested that gender-affirming hormone therapy (GAHT) may increase the risk of cardiovascular disease. However, owing to a paucity of research, the mechanisms underpinning these increased risks are poorly understood. Moreover, previous research has been limited by heterogenous methodologies, being underpowered, and lacking appropriate control populations. Consequently, the need for evidence regarding cardiovascular health in LGBTQ + individuals has been recognised as a critical area for future research to facilitate better healthcare and guidance. Recent research investigating the effect of transmasculine (testosterone) GAHT on cardiovascular disease risk points to testosterone effecting the nitric oxide pathway, triggering inflammation, and promoting endothelial dysfunction. Equivalent studies focussing on transfeminine (oestrogen) GAHT are required, representing a crucial area of future research. Furthermore, when examining the effects of GAHT on the vasculature, it cannot be ignored that there are multiple factors that may increase the burden of cardiovascular disease in the transgender population. Such stressors include major psychological stress; increased adverse health behaviours, such as smoking; discrimination; and lowered socioeconomic status; all of which undoubtedly impact upon cardiovascular disease risk and offers the opportunity for intervention.
Collapse
Affiliation(s)
- Charlotte N Murphy
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Eleanor Davies
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Paul J Connelly
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom.
| |
Collapse
|
16
|
Quintela-Castro FCDA, Pereira TSS, Alves DB, Chiepe L, Nascimento LS, Chiepe KCMB, Barcelos RM, Costa BM, Enriquez-Martinez OG, Rossoni JV, Bellettini-Santos T. Lipid profile and risk of cardiovascular disease in adult transgender men receiving cross-sex hormone therapy: a systematic review. Nutr Rev 2023; 81:1310-1320. [PMID: 36779324 DOI: 10.1093/nutrit/nuad003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
CONTEXT A recent US national survey of the health status of the male transgender population has raised awareness about the little-studied relationship between testosterone hormone therapy in transgender men and cardiovascular outcomes. OBJECTIVE The aim of this systematic review was to assess the relationship between cross-sex hormone therapy in transgender men and lipid profiles and cardiovascular risk. DATA SOURCES The PubMed, SciELO, SpringerLink, and EBSCOhost databases were searched up to March 2021 for studies assessing the association between cross-sex hormone therapy and the incidence of outcomes related to cardiovascular disease in transgender men over 18 years of age . DATA EXTRACTION Data extracted were sorted into clinical data (systolic, diastolic, and mean blood pressure), anthropometric data (body mass index, weight, waist circumference, fat mass, and lean mass), and biochemical data (triglycerides, total cholesterol, low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein cholesterol [HDL-C], very low-density lipoprotein cholesterol [VLDL-C], and the HDL-C to LDL-C ratio). DATA ANALYSIS Study quality was appraised independently by two reviewers using the Cochrane tools for assessment of methodological quality or risk of bias in nonrandomized studies, and the Newcastle-Ottawa Scale was applied. Of 735 studies identified, 11 were included in the review. Most studies reported no change in cholesterol or triglyceride levels after hormone treatment. A reduction in HDL-C levels was observed in 7 of 11 studies, although this alone cannot be considered a cardiovascular risk factor. Likewise, clinical and anthropometric findings showed no changes predictive of cardiovascular risk. CONCLUSIONS Although these findings suggest that hormone therapy may lead to a decrease in HDL-C levels and an increase in LDL-C levels, they are insufficient to establish a relationship with cardiovascular disease. Furthermore, no significant effects on metabolic and anthropometric values were found. Further studies with higher quality and longer follow-up periods are needed to establish cardiovascular risk. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration number CRD 42020212560.
Collapse
Affiliation(s)
| | | | - Danúbia Boy Alves
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| | - Letícia Chiepe
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| | - Laura Sperandio Nascimento
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| | | | - Rafael Mazioli Barcelos
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| | - Bruno Maia Costa
- Department of Health, Multivix College São Mateus, São Mateus, Espirito Santo, Brazil
| | | | - Joamyr Victor Rossoni
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| | - Tatiani Bellettini-Santos
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, Espirito Santo, Brazil
| |
Collapse
|
17
|
Reyes-García J, Díaz-Hernández V, Carbajal-García A, Casas-Hernández MF, Sommer B, Montaño LM. Theophylline-Induced Relaxation Is Enhanced after Testosterone Treatment via Increased K V1.2 and K V1.5 Protein Expression in Guinea Pig Tracheal Smooth Muscle. Int J Mol Sci 2023; 24:ijms24065884. [PMID: 36982957 PMCID: PMC10059212 DOI: 10.3390/ijms24065884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/16/2023] [Accepted: 02/25/2023] [Indexed: 03/30/2023] Open
Abstract
Theophylline is a drug commonly used to treat asthma due to its anti-inflammatory and bronchodilatory properties. Testosterone (TES) has been suggested to reduce the severity of asthma symptoms. This condition affects boys more than girls in childhood, and this ratio reverses at puberty. We reported that guinea pig tracheal tissue chronic exposure to TES increases the expression of β2-adrenoreceptors and enhances salbutamol-induced K+ currents (IK+). Herein, we investigated whether the upregulation of K+ channels can enhance the relaxation response to methylxanthines, including theophylline. Chronic incubation of guinea pig tracheas with TES (40 nM, 48 h) enhanced the relaxation induced by caffeine, isobutylmethylxanthine, and theophylline, an effect that was abolished by tetraethylammonium. In tracheal myocytes, chronic incubation with TES increased theophylline-induced IK+; flutamide reversed this effect. The increase in IK+ was blocked by 4-aminopyridine by ~82%, whereas iberiotoxin reduced IK+ by ~17%. Immunofluorescence studies showed that chronic TES exposure increased the expression of KV1.2 and KV1.5 in airway smooth muscle (ASM). In conclusion, chronic exposure to TES in guinea pig ASM promotes upregulation of KV1.2 and KV1.5 and enhances theophylline relaxation response. Therefore, gender should be considered when prescribing methylxanthines, as teenage boys and males are likely to respond better than females.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Verónica Díaz-Hernández
- Departamento de Embriología y Genética, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - María F Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City 14080, Mexico
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
18
|
Age-related decrease in serum dihydrotestosterone concentration is accompanied by impaired vascular status. Exp Gerontol 2023; 173:112104. [PMID: 36693531 DOI: 10.1016/j.exger.2023.112104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/03/2022] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
INTRODUCTION The effect of androgens on the cardiovascular system in humans is ambiguous. Moreover, still little is known about the effects of the most potent androgen, dihydrotestosterone, on arterial stiffness and endothelial function. The aim of this study was to evaluate whether age-dependent alterations in serum concentration of dihydrotestosterone and its circulating metabolite are accompanied by changes in endothelial function and arterial stiffness. METHODS In 12 young and 11 older men, basal serum concentrations of testosterone, dehydroepiandrosterone sulfate (DHAE-S), androstenedione (AE), dihydrotestosterone (DHT) and androstanediol glucuronide (ADG) were analyzed in relation to vascular status including cIMT - carotid intima media thickness, cAI - central augmentation index, crPWV - carotid radial pulse wave velocity, SI - stiffness index, endothelial and inflammatory markers. RESULTS Although concentration of testosterone was not different between young and older group, it was demonstrated that DHT, DHEA-S, AE and ADG were significantly lower in older men in comparison to young men (p < 0.01). Interestingly the most surprising difference was found for DHT concentration, that was as much as 61 % lower in aged men that displayed significantly higher values of cIMT, AI, crPWV and SI (p < 10-4), suggestive of arterial stiffness. Furthermore, DHT was negatively correlated to all arterial wall parameters (cAI, crPWV, SI and cIMT), c-reactive protein (CRP) and hyaluronic acid (HA) concentration, as well as positively correlated to markers of endothelial function (MNA and 6-keto-PGF1α) in all studied individuals (n = 23). CONCLUSIONS We have shown that ageing leads to a significant decrease in DHT concentration that is accompanied by impaired arterial wall characteristics and worsened endothelial function. Therefore more attention should be paid to the DHT, DHEA-S and ADG concentrations as a biomarkers for vascular dysfunction in ageing men.
Collapse
|
19
|
Mu X, Liu S, Wang Z, Jiang K, McClintock T, Stromberg AJ, Tezanos AV, Lee ES, Curci JA, Gong MC, Guo Z. Androgen aggravates aortic aneurysms via suppressing PD-1 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525073. [PMID: 36711644 PMCID: PMC9882344 DOI: 10.1101/2023.01.22.525073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Androgen has long been recognized for its pivotal role in the sexual dimorphism of cardiovascular diseases, including aortic aneurysms, a devastating vascular disease with a higher prevalence and mortality rate in men than women. However, the molecular mechanism by which androgen mediates aortic aneurysms is largely unknown. Here, we report that male but not female mice develop aortic aneurysms in response to aldosterone and high salt (Aldo-salt). We demonstrate that both androgen and androgen receptors (AR) are crucial for the sexually dimorphic response to Aldo-salt. We identify T cells expressing programmed cell death protein 1 (PD-1), an immune checkpoint molecule important in immunity and cancer immunotherapy, as a key link between androgen and aortic aneurysms. We show that intraperitoneal injection of anti-PD-1 antibody reinstates Aldo-salt-induced aortic aneurysms in orchiectomized mice. Mechanistically, we demonstrate that AR binds to the PD-1 promoter to suppress its expression in the spleen. Hence, our study reveals an important but unexplored mechanism by which androgen contributes to aortic aneurysms by suppressing PD-1 expression in T cells. Our study also suggests that cancer patients predisposed to the risk factors of aortic aneurysms may be advised to screen for aortic aneurysms during immune checkpoint therapy.
Collapse
|
20
|
Rouver WDN, Delgado NTB, Gonçalves LT, Giesen JAS, Santos da Costa C, Merlo E, Damasceno Costa E, Lemos VS, Bernardes Graceli J, Santos RLD. Sex hormones and vascular reactivity: a temporal evaluation in resistance arteries of male rats. J Mol Endocrinol 2023; 70:e220147. [PMID: 36476761 DOI: 10.1530/jme-22-0147] [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: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
The role of androgens in vascular reactivity is controversial, particularly regarding their age-related actions. The objective of this study was to conduct a temporal evaluation of the vascular reactivity of resistance arteries of young male rats, as well as to understand how male sex hormones can influence the vascular function of these animals. Endothelium-mediated relaxation was characterized in third-order mesenteric arteries of 10-, 12-, 16-, and 18w (week-old) male rats. Concentration-response curves to acetylcholine (ACh, 0.1 nmol/L-10 µmol/L) were constructed in arteries previously contracted with phenylephrine (PE, 3 µmol/L), before and after the use of nitric oxide synthase or cyclooxygenase inhibitors. PE concentration-response curves (1 nmol/L-100 μmol/L) were also built. The levels of vascular nitric oxide, superoxide anion, and hydrogen peroxide were assessed and histomorphometry analysis was performed. The 18w group had impaired endothelium-dependent relaxation. All groups showed prostanoid-independent and nitric oxide-dependent vasodilatory response, although this dependence seems to be smaller in the 18w group. The 18w group had the lowest nitric oxide and hydrogen peroxide production, in addition to the highest superoxide anion levels. Besides functional impairment, 18w animals showed morphological differences in third-order mesenteric arteries compared with the other groups. Our data show that time-dependent exposure to male sex hormones appears to play an important role in the development of vascular changes that can lead to impaired vascular reactivity in mesenteric arteries, which could be related to the onset of age-related cardiovascular changes in males.
Collapse
Affiliation(s)
- Wender do Nascimento Rouver
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Leticia Tinoco Gonçalves
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Charles Santos da Costa
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Merlo
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Damasceno Costa
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Virginia Soares Lemos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jones Bernardes Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| |
Collapse
|
21
|
Lawrence BM, O’Donnell L, Smith LB, Rebourcet D. New Insights into Testosterone Biosynthesis: Novel Observations from HSD17B3 Deficient Mice. Int J Mol Sci 2022; 23:ijms232415555. [PMID: 36555196 PMCID: PMC9779265 DOI: 10.3390/ijms232415555] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Androgens such as testosterone and dihydrotestosterone (DHT) are essential for male sexual development, masculinisation, and fertility. Testosterone is produced via the canonical androgen production pathway and is essential for normal masculinisation and testis function. Disruption to androgen production can result in disorders of sexual development (DSD). In the canonical pathway, 17β-hydroxysteroid dehydrogenase type 3 (HSD17B3) is viewed as a critical enzyme in the production of testosterone, performing the final conversion required. HSD17B3 deficiency in humans is associated with DSD due to low testosterone concentration during development. Individuals with HSD17B3 mutations have poorly masculinised external genitalia that can appear as ambiguous or female, whilst having internal Wolffian structures and testes. Recent studies in mice deficient in HSD17B3 have made the surprising finding that testosterone production is maintained, male mice are masculinised and remain fertile, suggesting differences between mice and human testosterone production exist. We discuss the phenotypic differences observed and the possible other pathways and enzymes that could be contributing to testosterone production and male development. The identification of alternative testosterone synthesising enzymes could inform the development of novel therapies to endogenously regulate testosterone production in individuals with testosterone deficiency.
Collapse
Affiliation(s)
- Ben M. Lawrence
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
- Correspondence: (B.M.L.); (D.R.)
| | - Liza O’Donnell
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Lee B. Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
- Office for Research, Griffith University, Southport, QLD 4222, Australia
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
- Correspondence: (B.M.L.); (D.R.)
| |
Collapse
|
22
|
Dahiya V, Bagchi G. Non-canonical androgen signaling pathways and implications in prostate cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119357. [PMID: 36100060 DOI: 10.1016/j.bbamcr.2022.119357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Androgen signaling is a critical determinant of timely and proper development of all male organs including the prostate. Maturation of prostate and its neoplastic transformation is intricately associated with accurate androgen signaling. Ablation of androgen has therefore been the primary treatment mechanism of Prostate cancer (PCa) patients for several decades. Upon removal, the tumor recedes for a while, yet it reappears soon, in an androgen independent state, untreatable by current therapeutic regimens. Studies reveal that apart from the classical androgen signaling pathway known and targeted for almost a century, there exist several non-canonical pathways, with marked impact on classical androgen signaling and PCa growth. These include non-genomic signaling by androgens via alternate membrane GPCRs, signaling by non-androgens that ultimately impact the androgen signaling pathway, or an integration of non-genomic and genomic response as seen in case of protein kinase A activation. Accurate understanding of these various non-canonical androgen signaling pathways and their influence on the typical androgen signaling pathway can help design important interventions for PCa patients. This review analyses in detail the various non-classical androgen signaling pathways and their impact, if any, on classical mode of androgen action and PCa.
Collapse
Affiliation(s)
- Versha Dahiya
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413
| | - Gargi Bagchi
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413.
| |
Collapse
|
23
|
Lucas-Herald AK, Touyz RM. Androgens and Androgen Receptors as Determinants of Vascular Sex Differences Across the Lifespan. Can J Cardiol 2022; 38:1854-1864. [PMID: 36156286 DOI: 10.1016/j.cjca.2022.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022] Open
Abstract
Androgens, including testosterone and its more potent metabolite dihydrotestosterone, exert multiple actions in the body. Physiologically, they play a critical role in male sex development. In addition, they influence vascular function, including arterial vasodilation and mediation of myogenic tone. Androgens are produced from 9 weeks' gestation in the human fetal testis, as well as in small amounts by the adrenal glands. Serum concentrations vary according to age and sex. The vasculature is a target for direct actions of androgens, which bind to various sex hormone receptors expressed in endothelial and vascular smooth muscle cells. Androgens exert both vasoprotective and vasoinjurious effects, depending on multiple factors including sex-specific effects of androgens, heterogeneity of the vascular endothelium, differential expression of androgen and sex hormone receptors in endothelial and vascular smooth muscle cells, and the chronicity of androgen administration. Long-term administration of androgens induces vasoconstriction and influences endothelial permeability, whereas acute administration may have opposite effects. At the cellular level, androgens stimulate endothelial cell production of nitric oxide and inhibit proinflammatory signalling pathways, inducing vasorelaxation and vasoprotection. However, androgens also activate endothelial production of vasoconstrictors and stimulate recruitment of endothelial progenitor cells. In humans, both androgen deficiency and androgen excess are associated with increased cardiovascular morbidity and mortality. This review discusses how androgens modulate vascular sex differences across the lifespan by considering the actions and production of androgens in both sexes and describes how cardiovascular risk is altered as levels of androgens change with aging.
Collapse
Affiliation(s)
- Angela K Lucas-Herald
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom.
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montréal, Québec, Canada.
| |
Collapse
|
24
|
Rosato E, Sciarra F, Anastasiadou E, Lenzi A, Venneri MA. Revisiting the physiological role of androgens in women. Expert Rev Endocrinol Metab 2022; 17:547-561. [PMID: 36352537 DOI: 10.1080/17446651.2022.2144834] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Extensive research underlines the critical functions of androgens in females. Nevertheless, the precise mechanisms of their action are poorly understood. Here, we review the existing literature regarding the physiological role of androgens in women throughout life. AREAS COVERED Several studies show that androgen receptors (ARs) are broadly expressed in numerous female tissues. They are essential for many physiological processes, including reproductive, sexual, cardiovascular, bone, muscle, and brain health. They are also involved in adipose tissue and liver function. Androgen levels change with the menstrual cycle and decrease in the first decades of life, independently of menopause. EXPERT OPINION To date, studies are limited by including small numbers of women, the difficulty of dosing androgens, and their cyclical variations. In particular, whether androgens play any significant role in regulating the establishment of pregnancy is poorly understood. The neural functions of ARs have also been investigated less thoroughly, although it is expressed at high levels in brain structures. Moreover, the mechanism underlying the decline of dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) with age is unclear. Other factors, including estrogen's effect on adrenal androgen production, reciprocal regulation of ARs, and non-classical effects of androgens, remain to be determined.
Collapse
Affiliation(s)
- Elena Rosato
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Eleni Anastasiadou
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
25
|
Rzeszotek S, Kolasa A, Pilutin A, Misiakiewicz-Has K, Sielatycka K, Wiszniewska B. The Interplay between Finasteride-Induced Androgen Imbalance, Endoplasmic Reticulum Stress, Oxidative Stress, and Liver Disorders in Paternal and Filial Generation. Biomedicines 2022; 10:2725. [PMID: 36359245 PMCID: PMC9687381 DOI: 10.3390/biomedicines10112725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/20/2023] Open
Abstract
Finasteride (Fin) causes androgen imbalance by inhibiting the conversion of testosterone (T) to its more active metabolite, dihydrotestosterone (DHT). Androgen receptors (AR) are present (e.g., in hepatocytes), which have well-developed endoplasmic reticulum (ERet). Cellular protein quality control is carried out by ERet in two paths: (i) unfolded protein response (UPR) and/or (ii) endoplasmic reticulum associated degradation (ERAD). ERet under continuous stress can generate changes in the UPR and can direct the cell on the pathway of life or death. It has been demonstrated that genes involved in ERet stress are among the genes controlled by androgens in some tissues. Oxidative stress is also one of the factors affecting the functions of ERet and androgens are one of the regulators of antioxidant enzyme activity. In this paper, we discuss/analyze a possible relationship between androgen imbalance in paternal generation with ERet stress and liver disorders in both paternal and filial generation. In our rat model, hyperglycemia and subsequent higher accumulation of hepatic glycogen were observed in all filial generation obtained from females fertilized by Fin-treated males (F1:Fin). Importantly, genes encoding enzymes involved in glucose and glycogen metabolism have been previously recognized among UPR targets.
Collapse
Affiliation(s)
- Sylwia Rzeszotek
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Anna Pilutin
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Kamila Misiakiewicz-Has
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Katarzyna Sielatycka
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Szczecin, 71-415 Szczecin, Poland
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| |
Collapse
|
26
|
Galindez SM, Keightley A, Koulen P. Differential distribution of steroid hormone signaling networks in the human choroid-retinal pigment epithelial complex. BMC Ophthalmol 2022; 22:406. [PMID: 36266625 PMCID: PMC9583547 DOI: 10.1186/s12886-022-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background The retinal pigment epithelium (RPE), a layer of pigmented cells that lies between the neurosensory retina and the underlying choroid, plays a critical role in maintaining the functional integrity of photoreceptor cells and in mediating communication between the neurosensory retina and choroid. Prior studies have demonstrated neurotrophic effects of select steroids that mitigate the development and progression of retinal degenerative diseases via an array of distinct mechanisms of action. Methods Here, we identified major steroid hormone signaling pathways and their key functional protein constituents controlling steroid hormone signaling, which are potentially involved in the mitigation or propagation of retinal degenerative processes, from human proteome datasets with respect to their relative abundances in the retinal periphery, macula, and fovea. Results Androgen, glucocorticoid, and progesterone signaling networks were identified and displayed differential distribution patterns within these three anatomically distinct regions of the choroid-retinal pigment epithelial complex. Classical and non-classical estrogen and mineralocorticoid receptors were not identified. Conclusion Identified differential distribution patterns suggest both selective susceptibility to chronic neurodegenerative disease processes, as well as potential substrates for drug target discovery and novel drug development focused on steroid signaling pathways in the choroid-RPE.
Collapse
Affiliation(s)
- Sydney M Galindez
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Andrew Keightley
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Peter Koulen
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA. .,Department of Biomedical Sciences, University of Missouri - Kansas City School of Medicine, Kansas City, MO, USA.
| |
Collapse
|
27
|
Venkatesh VS, Grossmann M, Zajac JD, Davey RA. The role of the androgen receptor in the pathogenesis of obesity and its utility as a target for obesity treatments. Obes Rev 2022; 23:e13429. [PMID: 35083843 PMCID: PMC9286619 DOI: 10.1111/obr.13429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/09/2022] [Accepted: 01/09/2022] [Indexed: 11/27/2022]
Abstract
Obesity is associated with hypothalamic-pituitary-testicular axis dysregulation in males. Here, we summarize recent evidence derived from clinical trials and studies in preclinical animal models regarding the role of androgen receptor (AR) signaling in the pathophysiology of males with obesity. We also discuss therapeutic strategies targeting the AR for the treatment of obesity and their limitations and provide insight into the future research necessary to advance this field.
Collapse
Affiliation(s)
- Varun S Venkatesh
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria
| | - Mathis Grossmann
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria.,Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria.,Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria
| |
Collapse
|
28
|
Alloatti G, Penna C, Comità S, Tullio F, Aragno M, Biasi F, Pagliaro P. Aging, sex and NLRP3 inflammasome in cardiac ischaemic disease. Vascul Pharmacol 2022; 145:107001. [PMID: 35623548 DOI: 10.1016/j.vph.2022.107001] [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: 03/29/2022] [Revised: 05/01/2022] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
Abstract
Experimentally, many strong cardioprotective treatments have been identified in different animal models of acute ischaemia/reperfusion injury (IRI) and coronary artery disease (CAD). However, the translation of these cardioprotective therapies for the benefit of the patients into the clinical scenario has been very disappointing. The reasons for this lack are certainly multiple. Indeed, many confounding factors we must deal in clinical reality, such as aging, sex and inflammatory processes are neglected in many experiments. Due to the pivotal role of aging, sex and inflammation in determining cardiac ischaemic disease, in this review, we take into account age as a modifier of tolerance to IRI in the two sexes, dissecting aging and myocardial reperfusion injury mechanisms and the sex differences in tolerance to IRI. Then we focus on the role of the gut microbiota and the NLRP3 inflammasome in myocardial IRI and on the possibility to consider NLRP3 inflammasome as a potential target in the treatment of CAD in relationship with age and sex. Finally, we consider the cardioprotective mechanisms and cardioprotective treatments during aging in the two sexes.
Collapse
Affiliation(s)
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano, 10043 Torino, TO, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
29
|
Paletta P, Bass N, Aspesi D, Choleris E. Sex Differences in Social Cognition. Curr Top Behav Neurosci 2022; 62:207-234. [PMID: 35604571 DOI: 10.1007/7854_2022_325] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In this review we explore the sex differences underlying various types of social cognition. Particular focus will be placed on the behaviors of social recognition, social learning, and aggression. Known similarities and differences between sexes in the expressions of these behaviors and the known brain regions where these behaviors are mediated are discussed. The role that the sex hormones (estrogens and androgens) have as well as possible interactions with other neurochemicals, such as oxytocin, vasopressin, and serotonin is reviewed as well. Finally, implications about these findings on the mediation of social cognition are mediated and the sex differences related to humans are considered.
Collapse
Affiliation(s)
- Pietro Paletta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Noah Bass
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
30
|
Lucas-Herald AK, Montezano AC, Alves-Lopes R, Haddow L, Alimussina M, O’Toole S, Flett M, Lee B, Amjad SB, Steven M, Brooksbank K, McCallum L, Delles C, Padmanabhan S, Ahmed SF, Touyz RM. Vascular dysfunction and increased cardiovascular risk in hypospadias. Eur Heart J 2022; 43:1832-1845. [PMID: 35567552 PMCID: PMC9113289 DOI: 10.1093/eurheartj/ehac112] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/30/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
AIMS Hypogonadism is associated with cardiovascular disease. However, the cardiovascular impact of hypogonadism during development is unknown. Using hypospadias as a surrogate of hypogonadism, we investigated whether hypospadias is associated with vascular dysfunction and is a risk factor for cardiovascular disease. METHODS AND RESULTS Our human study spanned molecular mechanistic to epidemiological investigations. Clinical vascular phenotyping was performed in adolescents with hypospadias and controls. Small subcutaneous arteries from penile skin from boys undergoing hypospadias repair and controls were isolated and functional studies were assessed by myography. Vascular smooth muscle cells were used to assess: Rho kinase, reactive oxygen species (ROS), nitric oxide synthase/nitric oxide, and DNA damage. Systemic oxidative stress was assessed in plasma and urine. Hospital episode data compared men with a history of hypospadias vs. controls. In adolescents with hypospadias, systolic blood pressure (P = 0.005), pulse pressure (P = 0.03), and carotid intima-media thickness standard deviation scores (P = 0.01) were increased. Arteries from boys with hypospadias demonstrated increased U46619-induced vasoconstriction (P = 0.009) and reduced acetylcholine-induced endothelium-dependent (P < 0.0001) and sodium nitroprusside-induced endothelium-independent vasorelaxation (P < 0.0001). Men born with hypospadias were at increased risk of arrhythmia [odds ratio (OR) 2.8, 95% confidence interval (CI) 1.4-5.6, P = 0.003]; hypertension (OR 4.2, 95% CI 1.5-11.9, P = 0.04); and heart failure (OR 1.9, 95% CI 1.7-114.3, P = 0.02). CONCLUSION Hypospadias is associated with vascular dysfunction and predisposes to hypertension and cardiovascular disease in adulthood. Underlying mechanisms involve perturbed Rho kinase- and Nox5/ROS-dependent signalling. Our novel findings delineate molecular mechanisms of vascular injury in hypogonadism, and identify hypospadias as a cardiovascular risk factor in males.
Collapse
Affiliation(s)
- Angela K Lucas-Herald
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Rheure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Laura Haddow
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Malika Alimussina
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Stuart O’Toole
- Department of Pediatric Surgery, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Martyn Flett
- Department of Pediatric Surgery, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Boma Lee
- Department of Pediatric Surgery, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - S Basith Amjad
- Department of Pediatric Surgery, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Mairi Steven
- Department of Pediatric Surgery, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Katriona Brooksbank
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Linsay McCallum
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G45 8TF, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Centre for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| |
Collapse
|
31
|
Leciejewska N, Kołodziejski PA, Sassek M, Nogowski L, Małek E, Pruszyńska-Oszmałek E. Ostarine-Induced Myogenic Differentiation in C2C12, L6, and Rat Muscles. Int J Mol Sci 2022; 23:ijms23084404. [PMID: 35457222 PMCID: PMC9031805 DOI: 10.3390/ijms23084404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
Ostarine (also known as enobosarm or Gtx-024) belongs to the selective androgen receptor modulators (SARMs). It is a substance with an aryl-propionamide structure, classified as a non-steroidal compound that is not subjected to the typical steroid transformations of aromatization and reduction by α5 reductase. Despite ongoing research on ostarine, knowledge about it is still limited. Earlier studies indicated that ostarine may affect the metabolism of muscle tissue, but this mechanism has not been yet described. We aimed to investigate the effect of ostarine on the differentiation and metabolism of muscle. Using C2C12 and L6 cells, as well as muscles obtained from rats administered ostarine, we showed that ostarine stimulates C2C12 and L6 proliferation and cell viability and that this effect is mediated by androgen receptor (AR) and ERK1/2 kinase activation (p < 0.01). We also found that ostarine stimulates muscle cell differentiation by increasing myogenin, MyoD, and MyH expression in both types of cells (p < 0.01). Moreover, pharmacological blocking of AR inhibits the stimulatory effect of ostarine. We further demonstrated that 30 days of ostarine administration increases myogenin, MyoD, and MyH expression, as well as muscle mass, in rats (p < 0.01). Based on our research, we conclude that ostarine stimulates muscle tissue proliferation and differentiation via the androgen receptor.
Collapse
Affiliation(s)
- Natalia Leciejewska
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (N.L.); (P.A.K.); (M.S.); (L.N.)
| | - Paweł A. Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (N.L.); (P.A.K.); (M.S.); (L.N.)
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (N.L.); (P.A.K.); (M.S.); (L.N.)
| | - Leszek Nogowski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (N.L.); (P.A.K.); (M.S.); (L.N.)
| | - Emilian Małek
- Department of Preclinical Sciences and Infectious Diseases, Faculty of Veterinary Medicine and Animal Science, University of Life Sciences, 60-637 Poznan, Poland;
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (N.L.); (P.A.K.); (M.S.); (L.N.)
- Correspondence: ; Tel.: +48-61-8466084
| |
Collapse
|
32
|
Abstract
Oxidative stress is caused by homeostasis disrupted by excessively increased reactive oxygen species (ROS) due to intrinsic or extrinsic causes. Among diseases caused by the abnormal induction of ROS, cancer is a representative disease that shows gender specificity in the development and malignancy. Females have the advantage of longer life expectancy than males because of the genetic advantages derived from X chromosomes, the antioxidant protective function by estrogen, and the decrease in exposure to extrinsic risk factors such as alcohol and smoking. This study first examines the ordinary biological responses to oxidative stress and the effects of ROS on the cancer progression and describes the differences in cancer incidence and mortality by gender and the differences in oxidative stress affected by sex hormones. This paper summarized how several important transcription factors regulate ROS-induced stress and in vivo responses, and how their expression is changed by sex hormones. Estrogen is associated with disease resistance and greater mitochondrial function, and reduces mitochondrial damage and ROS production in females than in males. In addition, estrogen affects the activation of nuclear factor-erythroid 2 p45-related factor (NRF) 2 and the regulation of other antioxidant-related transcription factors through NRF2, leading to benefits in females. Because ROS have a variety of molecular targets in cells, the effective cancer treatment requires understanding the potential of ROS and focusing on the characteristics of the research target such as patient's gender. Therefore, this review intends to emphasize the necessity of discussing gender specificity as a new therapeutic approach for efficient regulation of ROS considering individual specificity.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Chemistry, College of Science and Technology, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
33
|
Koukoulis GN, Filiponi M, Gougoura S, Befani C, Liakos P, Bargiota Α. Testosterone and dihydrotestosterone modulate the redox homeostasis of endothelium. Cell Biol Int 2022; 46:660-670. [DOI: 10.1002/cbin.11768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 12/26/2021] [Accepted: 01/02/2022] [Indexed: 12/08/2022]
Affiliation(s)
- George N Koukoulis
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Maria Filiponi
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Sofia Gougoura
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Christina Befani
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Panagiotis Liakos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Αlexandra Bargiota
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| |
Collapse
|
34
|
Could Lower Testosterone in Older Men Explain Higher COVID-19 Morbidity and Mortalities? Int J Mol Sci 2022; 23:ijms23020935. [PMID: 35055119 PMCID: PMC8781054 DOI: 10.3390/ijms23020935] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 01/08/2023] Open
Abstract
The health scourge imposed on humanity by the COVID-19 pandemic seems not to recede. This fact warrants refined and novel ideas analyzing different aspects of the illness. One such aspect is related to the observation that most COVID-19 casualties were older males, a tendency also noticed in the epidemics of SARS-CoV in 2003 and the Middle East respiratory syndrome in 2012. This gender-related difference in the COVID-19 death toll might be directly involved with testosterone (TEST) and its plasmatic concentration in men. TEST has been demonstrated to provide men with anti-inflammatory and immunological advantages. As the plasmatic concentration of this androgen decreases with age, the health benefit it confers also diminishes. Low plasmatic levels of TEST can be determinant in the infection’s outcome and might be related to a dysfunctional cell Ca2+ homeostasis. Not only does TEST modulate the activity of diverse proteins that regulate cellular calcium concentrations, but these proteins have also been proven to be necessary for the replication of many viruses. Therefore, we discuss herein how TEST regulates different Ca2+-handling proteins in healthy tissues and propose how low TEST concentrations might facilitate the replication of the SARS-CoV-2 virus through the lack of modulation of the mechanisms that regulate intracellular Ca2+ concentrations.
Collapse
|
35
|
Hashimoto D, Fujimoto K, Morioka S, Ayabe S, Kataoka T, Fukumura R, Ueda Y, Kajimoto M, Hyuga T, Suzuki K, Hara I, Asamura S, Wakana S, Yoshiki A, Gondo Y, Tamura M, Sasaki T, Yamada G. Establishment of mouse line showing inducible priapism-like phenotypes. Reprod Med Biol 2022; 21:e12472. [PMID: 35765371 PMCID: PMC9207557 DOI: 10.1002/rmb2.12472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/11/2022] Open
Abstract
Purpose Penile research is expected to reveal new targets for treatment and prevention of the complex mechanisms of its disorder including erectile dysfunction (ED). Thus, analyses of the molecular processes of penile ED and continuous erection as priapism are essential issues of reproductive medicine. Methods By performing mouse N-ethyl-N-nitrosourea mutagenesis and exome sequencing, we established a novel mouse line displaying protruded genitalia phenotype (PGP; priapism-like phenotype) and identified a novel Pitpna gene mutation for PGP. Extensive histological analyses on the Pitpna mutant and intracavernous pressure measurement (ICP) and liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI/MS)/MS analyses were performed. Results We evaluated the role of phospholipids during erection for the first time and showed the mutants of inducible phenotypes of priapism. Moreover, quantitative analysis using LC-ESI/MS/MS revealed that the level of phosphatidylinositol (PI) was significantly lower in the mutant penile samples. These results imply that PI may contribute to penile erection by PITPα. Conclusions Our findings suggest that the current mutant is a mouse model for priapism and abnormalities in PI signaling pathways through PITPα may lead to priapism providing an attractive novel therapeutic target in its treatment.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shin Morioka
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Shinya Ayabe
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Kataoka
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Ryutaro Fukumura
- Clinical Laboratories Department sSRL & Shizuoka Cancer Center Collaborative Laboratories, IncShizuoka PrefJapan
| | - Yuko Ueda
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Mizuki Kajimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Taiju Hyuga
- Department of Pediatric UrologyChildren's Medical Center TochigiJichi Medical UniversityTochigiJapan
| | - Kentaro Suzuki
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Isao Hara
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shigeharu Wakana
- Department of Animal ExperimentationFoundation for Biomedical Research and Innovation at KobeCreative Lab for Innovation in Kobe 5F 6‐3‐7KobeHyogoJapan
| | - Atsushi Yoshiki
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Yoichi Gondo
- Department of Molecular Life SciencesDivision of Basic Medical Science and Molecular MedicineTokai University School of MedicineIsehara‐shiKanagawaJapan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype AnalysisRIKEN BioResource Research CenterTsukubaIbarakiJapan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Gen Yamada
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
36
|
Converse A, Thomas P. Androgens promote vascular endothelial cell proliferation through activation of a ZIP9-dependent inhibitory G protein/PI3K-Akt/Erk/cyclin D1 pathway. Mol Cell Endocrinol 2021; 538:111461. [PMID: 34555425 DOI: 10.1016/j.mce.2021.111461] [Citation(s) in RCA: 9] [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: 05/16/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022]
Abstract
While androgens have been reported to mediate cardiovascular endothelial cell proliferation, the potential involvement of membrane androgen receptors (mAR) has not been examined. Here we show ZIP9, a recently characterized mAR, mediates androgen-induced early proliferative events in human umbilical vein endothelial cells (HUVECs). Androgen treatment significantly increased cyclin D1 nuclear localization and proliferation, which were blocked by transfection with siRNA targeting ZIP9 but not the nuclear AR. Testosterone rapidly activated inhibitory G protein signaling, Erk, and Akt, and inhibition of these signaling members abrogated the ZIP9-mediated cyclin D1 and proliferative responses. Erk and Akt modulated cyclin D1 nuclear localization by upregulation of cyclin D1 mRNA and inhibition of GSK-3β activity, respectively. This is the first study to demonstrate a role for ZIP9 in HUVEC proliferation and indicates ZIP9 is a physiologically-relevant androgen receptor in the cardiovascular system that merits further study as a potential therapeutic target for treating cardiovascular disease.
Collapse
Affiliation(s)
- Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, Port Aransas, TX, USA.
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, Port Aransas, TX, USA
| |
Collapse
|
37
|
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
|
38
|
Haptoglobin genotypes and risk of vascular complications in a northern Chinese Han population with type 2 diabetes mellitus. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-021-01002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Revealing the Influences of Sex Hormones and Sex Differences in Atrial Fibrillation and Vascular Cognitive Impairment. Int J Mol Sci 2021; 22:ijms22168776. [PMID: 34445515 PMCID: PMC8396287 DOI: 10.3390/ijms22168776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
The impacts of sex differences on the biology of various organ systems and the influences of sex hormones on modulating health and disease have become increasingly relevant in clinical and biomedical research. A growing body of evidence has recently suggested fundamental sex differences in cardiovascular and cognitive function, including anatomy, pathophysiology, incidence and age of disease onset, symptoms affecting disease diagnosis, disease severity, progression, and treatment responses and outcomes. Atrial fibrillation (AF) is currently recognized as the most prevalent sustained arrhythmia and might contribute to the pathogenesis and progression of vascular cognitive impairment (VCI), including a range of cognitive deficits, from mild cognitive impairment to dementia. In this review, we describe sex-based differences and sex hormone functions in the physiology of the brain and vasculature and the pathophysiology of disorders therein, with special emphasis on AF and VCI. Deciphering how sex hormones and their receptor signaling (estrogen and androgen receptors) potentially impact on sex differences could help to reveal disease links between AF and VCI and identify therapeutic targets that may lead to potentially novel therapeutic interventions early in the disease course of AF and VCI.
Collapse
|
40
|
Lim WF, Forouhan M, Roberts TC, Dabney J, Ellerington R, Speciale AA, Manzano R, Lieto M, Sangha G, Banerjee S, Conceição M, Cravo L, Biscans A, Roux L, Pourshafie N, Grunseich C, Duguez S, Khvorova A, Pennuto M, Cortes CJ, La Spada AR, Fischbeck KH, Wood MJA, Rinaldi C. Gene therapy with AR isoform 2 rescues spinal and bulbar muscular atrophy phenotype by modulating AR transcriptional activity. SCIENCE ADVANCES 2021; 7:7/34/eabi6896. [PMID: 34417184 PMCID: PMC8378820 DOI: 10.1126/sciadv.abi6896] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is an X-linked, adult-onset neuromuscular condition caused by an abnormal polyglutamine (polyQ) tract expansion in androgen receptor (AR) protein. SBMA is a disease with high unmet clinical need. Recent studies have shown that mutant AR-altered transcriptional activity is key to disease pathogenesis. Restoring the transcriptional dysregulation without affecting other AR critical functions holds great promise for the treatment of SBMA and other AR-related conditions; however, how this targeted approach can be achieved and translated into a clinical application remains to be understood. Here, we characterized the role of AR isoform 2, a naturally occurring variant encoding a truncated AR lacking the polyQ-harboring domain, as a regulatory switch of AR genomic functions in androgen-responsive tissues. Delivery of this isoform using a recombinant adeno-associated virus vector type 9 resulted in amelioration of the disease phenotype in SBMA mice by restoring polyQ AR-dysregulated transcriptional activity.
Collapse
Affiliation(s)
- Wooi F Lim
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Mitra Forouhan
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Jesse Dabney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | - Raquel Manzano
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Maria Lieto
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Gavinda Sangha
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Subhashis Banerjee
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Lara Cravo
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Annabelle Biscans
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Loïc Roux
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Naemeh Pourshafie
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA
| | - Stephanie Duguez
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Londonderry, UK
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Maria Pennuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Constanza J Cortes
- Department of Neurology, Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC, USA
| | - Albert R La Spada
- Departments of Pathology and Laboratory Medicine, Neurology, and Biological Chemistry and the UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD, USA
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Carlo Rinaldi
- Department of Paediatrics, University of Oxford, Oxford, UK.
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Cittadini A, Isidori AM, Salzano A. Testosterone therapy and cardiovascular diseases. Cardiovasc Res 2021; 118:2039-2057. [PMID: 34293112 DOI: 10.1093/cvr/cvab241] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/16/2021] [Indexed: 11/14/2022] Open
Abstract
Since it was first synthesised in 1935, testosterone (T) has been viewed as the mythical Fountain of Youth, promising rejuvenation, restoring sexual appetites, growing stronger muscles, and quicker thinking. T is endowed with direct effects on myocardial and vascular structure and function, as well as on risk factors for cardiovascular (CV) disease. Indeed, low serum T levels are a risk factor for diabetes, metabolic syndrome, inflammation, and dyslipidaemia. Moreover, many studies have shown that T deficiency per se is an independent risk factor of CV and all-cause mortality. On this background and due to direct-to-patient marketing by drug companies, we have witnessed to the widespread use of T replacement therapy (TT) without clear indications particularly in late-life onset hypogonadism. The current review will dwell upon current evidence and controversies surrounding the role of T in the pathophysiology of CV diseases, the link between circulating T levels and CV risk, and the use of replacing T as a possible adjuvant treatment in specific CV disorders. Specifically, recent findings suggest that heart failure and type 2 diabetes mellitus represent two potential targets of T therapy once that a state of hypogonadism is diagnosed. However, only if ongoing studies solve the CV safety issue the T orchid may eventually 'bloom'.
Collapse
Affiliation(s)
- Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Interdisciplinary Research Centre on Biomaterials (CRIB), Federico II University, Naples, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Salzano
- IRCCS SDN, Diagnostic and Nuclear Research Institute, Naples, Italy
| |
Collapse
|
42
|
Pan T, Lin SC, Lee YC, Yu G, Song JH, Pan J, Titus M, Satcher RL, Panaretakis T, Logothetis C, Yu-Lee LY, Lin SH. Statins reduce castration-induced bone marrow adiposity and prostate cancer progression in bone. Oncogene 2021; 40:4592-4603. [PMID: 34127814 PMCID: PMC8384136 DOI: 10.1038/s41388-021-01874-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/14/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
A fraction of patients undergoing androgen deprivation therapy (ADT) for advanced prostate cancer (PCa) will develop recurrent castrate-resistant PCa (CRPC) in bone. Strategies to prevent CRPC relapse in bone are lacking. Here we show that the cholesterol-lowering drugs statins decrease castration-induced bone marrow adiposity in the tumor microenvironment and reduce PCa progression in bone. Using primary bone marrow stromal cells (BMSC) and M2-10B4 cells, we showed that ADT increases bone marrow adiposity by enhancing BMSC-to-adipocyte transition in vitro. Knockdown of androgen receptor abrogated BMSC-to-adipocyte transition, suggesting an androgen receptor-dependent event. RNAseq analysis showed that androgens reduce the secretion of adipocyte hormones/cytokines including leptin during BMSC-to-adipocyte transition. Treatment of PCa C4-2b, C4-2B4, and PC3 cells with leptin led to an increase in cell cycle progression and nuclear Stat3. RNAseq analysis also showed that androgens inhibit cholesterol biosynthesis pathway, raising the possibility that inhibiting cholesterol biosynthesis may decrease BMSC-to-adipocyte transition. Indeed, statins decreased BMSC-to-adipocyte transition in vitro and castration-induced bone marrow adiposity in vivo. Statin pre-treatment reduced 22RV1 PCa progression in bone after ADT. Our findings with statin may provide one of the mechanisms to the clinical correlations that statin use in patients undergoing ADT seems to delay progression to "lethal" PCa.
Collapse
Affiliation(s)
- Tianhong Pan
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Guoyu Yu
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Pan
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Satcher
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
Pillerová M, Borbélyová V, Hodosy J, Riljak V, Renczés E, Frick KM, Tóthová Ľ. On the role of sex steroids in biological functions by classical and non-classical pathways. An update. Front Neuroendocrinol 2021; 62:100926. [PMID: 34089761 PMCID: PMC8523217 DOI: 10.1016/j.yfrne.2021.100926] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/02/2022]
Abstract
The sex steroid hormones (SSHs) play several roles in regulation of various processes in the cardiovascular, immune, muscular and neural systems. SSHs affect prenatal and postnatal development of various brain structures, including regions associated with important physiological, behavioral, cognitive, and emotional functions. This action can be mediated by either intracellular or transmembrane receptors. While the classical mechanisms of SSHs action are relatively well examined, the physiological importance of non-classical mechanism of SSHs action through membrane-associated and transmembrane receptors in the brain remains unclear. The most recent summary describing the role of SSHs in different body systems is lacking. Therefore, the aim of this review is to discuss classical and non-classical signaling pathways of testosterone and estradiol action via their receptors at functional, cellular, tissue level and to describe the effects on various body systems and behavior. Particular emphasis will be on brain regions including the hippocampus, hypothalamus, frontal cortex and cerebellum.
Collapse
Affiliation(s)
- Miriam Pillerová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Július Hodosy
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Vladimír Riljak
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Emese Renczés
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ľubomíra Tóthová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia.
| |
Collapse
|
44
|
Banga S, Heinze-Milne SD, Godin J, Howlett SE. Signs of diastolic dysfunction are graded by serum testosterone levels in aging C57BL/6 male mice. Mech Ageing Dev 2021; 198:111523. [PMID: 34166687 DOI: 10.1016/j.mad.2021.111523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023]
Abstract
We investigated whether maladaptive, age-associated changes in heart structure and function were linked to circulating testosterone levels. Male C57BL/6 mice had a gonadectomy (GDX) or sham surgery at 4 weeks and effects of GDX on the heart were examined with echocardiography. Serum testosterone was measured with ELISA. Left ventricular (LV) mass increased with age but was smaller in GDX mice than sham at 18 months (144.0 ± 8.7 vs 118.2 ± 11.9 mg; p = 0.009). The isovolumic relaxation time (IVRT) declined with age but was prolonged in GDX mice at 18 months (10.5 ± 0.8 vs 12.5 ± 0.5 msec, p = 0.008). Ejection fraction did not change with age or GDX, but E/A ratios were lower in GDX mice than controls at 18 months (1.6 ± 0.2 vs 1.3 ± 0.1, p = 0.021). When links between serum testosterone and cardiac parameters were examined longitudinally in 18-24-month-old mice, LV mass declined with decreasing testosterone (β = 37.70, p = 0.016), however IVRT increased as testosterone decreased (β=-2.69, p = 0.036). Since longer IVRT and lower E/A ratios are signs of diastolic dysfunction, low circulating testosterone may promote or exacerbate diastolic dysfunction in older males. These findings suggest that lower testosterone directly modifies heart structure and function to promote maladaptive remodeling and diastolic dysfunction in the aging heart.
Collapse
Affiliation(s)
- Shubham Banga
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.
| | | | - Judith Godin
- Geriatric Medicine Research, Division of Geriatric Medicine, Nova Scotia Health Authority and Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada; Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
45
|
Sgrò P, Minganti C, Lista M, Antinozzi C, Cappa M, Pitsiladis Y, Pigozzi F, Di Luigi L. Dihydrotestosterone (DHT) rapidly increase after maximal aerobic exercise in healthy males: the lowering effect of phosphodiesterase's type 5 inhibitors on DHT response to exercise-related stress. J Endocrinol Invest 2021; 44:1219-1228. [PMID: 32946077 DOI: 10.1007/s40618-020-01409-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/26/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Few data exist on dihydrotestosterone (DHT) adaptation to exercise-related stress. The aim of the study was to investigate on serum DHT and other androgens' responses to acute aerobic exercises, and to verify if a long-acting phosphodiesterase's type 5 inhibitors could influence these responses, as previously observed for salivary testosterone. METHODS In a double-blind cross over study, 12 healthy trained male volunteers were submitted to both an acute sub-maximal and maximal exercise tests on cycle ergometer, after randomly receiving a two days placebo or tadalafil administration (20 mg, Cialis®, Ely-Lilly, Indianapolis, IN, USA). Blood sample collections were performed at different time points before and after exercise. Serum DHT, total testosterone (TT), dehydroepiandrosterone sulfate (DHEAS) and luteinizing hormone (LH), were assayed. RESULTS Serum DHT increase in placebo treatment immediately post maximal aerobic exercise and return to basal values at 60 min of recovery whereas tadalafil administration significantly reduced the DHT increase after exercise. The values of areas under curves showed the increase of TT after acute sub-maximal and maximal exercise and of DHEAS only after acute maximal aerobic exercise independently from treatment. CONCLUSIONS In addition to testosterone, also DHT plays an exercise-related adaptive role during high intensity aerobic exercise, but its rapid useful effects during exercise have to be determined. We hypothesized that the increased androgens secretion during exercise could be mainly related to steroidogenic enzymes modifications in peripheral tissues (i.e., muscles). Moreover, the blunting effect of tadalafil on DHT increase support a possible role of peripheral nitric oxide/GMPc related pathways in influencing physical-stress related DHT metabolism.
Collapse
Affiliation(s)
- P Sgrò
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Piazza Lauro de Bosis 15, 00135, Roma, Italy.
| | - C Minganti
- Unit of Sport Medicine, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Roma, Italy
| | - M Lista
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Piazza Lauro de Bosis 15, 00135, Roma, Italy
| | - C Antinozzi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Piazza Lauro de Bosis 15, 00135, Roma, Italy
| | - M Cappa
- Unit of Endocrinology, Bambino Gesù Children's Hospital, Roma, Italy
| | - Y Pitsiladis
- Collaborating Centre of Sports Medicine, University of Brighton, Welkin House, Eastbourne, UK
| | - F Pigozzi
- Unit of Sport Medicine, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Roma, Italy
| | - L Di Luigi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Piazza Lauro de Bosis 15, 00135, Roma, Italy
| |
Collapse
|
46
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
47
|
Querio G, Geddo F, Antoniotti S, Gallo MP, Penna C. Sex and Response to Cardioprotective Conditioning Maneuvers. Front Physiol 2021; 12:667961. [PMID: 34054579 PMCID: PMC8160310 DOI: 10.3389/fphys.2021.667961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/30/2021] [Indexed: 12/02/2022] Open
Abstract
Ischemic heart disease (IHD) is a multifactorial pathological condition strictly related to genetic, dietary, and lifestyle factors. Its morbidity and mortality rate represent one of the most important pathological issues that today involve younger people in a stronger way than in the past. IHD clinical outcomes are difficult to treat and have a high economic impact on health care. So prevention of this pathological condition through cardioprotective maneuvers represents the first line of intervention, as already underlined by several animal and human studies. Even if the time of intervention is important to prevent severe outcomes, many studies highlight that sex-dependent responses are crucial for the result of cardioprotective procedures. In this scenario sexual hormones have revealed an important role in cardioprotective approach, as women seem to be more protected toward cardiac insults when compared to male counterparts. The aim of this mini review is to show the molecular pathways involved in cardioprotective protocols and to elucidate how sexual hormones can contribute in ameliorating or worsening the physiological responses to IHD.
Collapse
Affiliation(s)
- Giulia Querio
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Federica Geddo
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
48
|
Fontányi Z, Sziva RE, Pál É, Hadjadj L, Monori-Kiss A, Horváth EM, Benkő R, Magyar A, Heinzlmann A, Benyó Z, Nádasy GL, Masszi G, Várbíró S. Vitamin D Deficiency Reduces Vascular Reactivity of Coronary Arterioles in Male Rats. Curr Issues Mol Biol 2021; 43:79-92. [PMID: 34066967 PMCID: PMC8928984 DOI: 10.3390/cimb43010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Vitamin D deficiency (VDD) may be considered an independent cardiovascular (CV) risk factor, and it is well known that CV risk is higher in males. Our goal was to investigate the pharmacological reactivity and receptor expression of intramural coronary artery segments of male rats in cases of different vitamin D supply. METHODS Four-week-old male Wistar rats were divided into a control group (n = 11) with optimal vitamin D supply (300 IU/kgbw/day) and a VDD group (n = 11, <0.5 IU/kgbw/day). After 8 weeks of treatment, intramural coronary artery segments were microprepared, their pharmacological reactivity was examined by in vitro microangiometry, and their receptor expression was investigated by immunohistochemistry. RESULTS Thromboxane A2 (TXA2)-agonist induced reduced vasoconstriction, testosterone (T) and 17-β-estradiol (E2) relaxations were significantly decreased, a significant decrease in thromboxane receptor (TP) expression was shown, and the reduction in estrogen receptor-α (ERα) expression was on the border of significance in the VDD group. CONCLUSIONS VD-deficient male coronary arteries showed deteriorated pharmacological reactivity to TXA2 and sexual steroids (E2, T). Insufficient vasoconstrictor capacity was accompanied by decreased TP receptor expression, and vasodilator impairments were mainly functional. The decrease in vasoconstrictor and vasodilator responses results in narrowed adaptational range of coronaries, causing inadequate coronary perfusion that might contribute to the increased CV risk in VDD.
Collapse
Affiliation(s)
- Zoltán Fontányi
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (Z.F.); (S.V.)
| | - Réka Eszter Sziva
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (Z.F.); (S.V.)
- Department of Physiology, Semmelweis University, Tűzoltó Street 37-47, 1094 Budapest, Hungary; (E.M.H.); (R.B.); (G.L.N.)
| | - Éva Pál
- Department of Translational Medicine, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (É.P.); (L.H.); (A.M.-K.); (Z.B.)
| | - Leila Hadjadj
- Department of Translational Medicine, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (É.P.); (L.H.); (A.M.-K.); (Z.B.)
| | - Anna Monori-Kiss
- Department of Translational Medicine, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (É.P.); (L.H.); (A.M.-K.); (Z.B.)
| | - Eszter Mária Horváth
- Department of Physiology, Semmelweis University, Tűzoltó Street 37-47, 1094 Budapest, Hungary; (E.M.H.); (R.B.); (G.L.N.)
| | - Rita Benkő
- Department of Physiology, Semmelweis University, Tűzoltó Street 37-47, 1094 Budapest, Hungary; (E.M.H.); (R.B.); (G.L.N.)
| | - Attila Magyar
- Department of Anatomy, Histology and Embriology, Semmelweis University, Tűzoltó Street 58, 1094 Budapest, Hungary;
| | - Andrea Heinzlmann
- Department of Anatomy and Histology, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary;
| | - Zoltán Benyó
- Department of Translational Medicine, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (É.P.); (L.H.); (A.M.-K.); (Z.B.)
| | - György L. Nádasy
- Department of Physiology, Semmelweis University, Tűzoltó Street 37-47, 1094 Budapest, Hungary; (E.M.H.); (R.B.); (G.L.N.)
| | - Gabriella Masszi
- Department of Internal Medicine, National Institute of Mental Health, Neurology and Neurosurgery, Lehel Street 59-61, 1135 Budapest, Hungary;
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynaecology, Semmelweis University, Üllői Street 78/a, 1082 Budapest, Hungary; (Z.F.); (S.V.)
| |
Collapse
|
49
|
Connelly PJ, Clark A, Touyz RM, Delles C. Transgender adults, gender-affirming hormone therapy and blood pressure: a systematic review. J Hypertens 2021; 39:223-230. [PMID: 32809982 PMCID: PMC7810409 DOI: 10.1097/hjh.0000000000002632] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Gender-affirming hormone therapy (GHT) is utilized by people who are transgender to align their secondary sex characteristics with their gender identity. Data relating to cardiovascular outcomes in this population are limited. We aimed to review the impact of GHT on the blood pressure (BP) of transgender individuals. METHODS We searched PubMed/MEDLINE, SCOPUS and Cochrane Library databases for articles published relating to the BP of transgender adults commencing GHT. Methodological quality was assessed via the 'Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group'. RESULTS Six hundred articles were screened, of which 14 studies were included in this systematic review encompassing 1309 individuals (∼50% transgender men and women) treated with GHT between 1989 and 2019. These articles were all pre-post observational studies without control groups. Mean ages ranged between 23.0-36.7 years (transgender men) and 25.2-34.8 years (transgender women). Interventions were diverse and included oral, transdermal and injectable hormonal preparations with 4 months to 5 years follow-up. Most studies in transgender men did not demonstrate a change in BP, whereas transgender women on GHT demonstrated both increases and decreases in SBP. These studies were heterogenous with significant methodological limitations and only two were determined to have a good quality rating. CONCLUSION There is currently insufficient data to advise the impact of GHT on BP in transgender individuals. Better quality research is essential to elucidate whether exogenous sex hormones modulate BP in transgender people and whether this putative alteration infers poorer cardiovascular outcomes.
Collapse
Affiliation(s)
- Paul J Connelly
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
50
|
Shah W, Khan R, Shah B, Khan A, Dil S, Liu W, Wen J, Jiang X. The Molecular Mechanism of Sex Hormones on Sertoli Cell Development and Proliferation. Front Endocrinol (Lausanne) 2021; 12:648141. [PMID: 34367061 PMCID: PMC8344352 DOI: 10.3389/fendo.2021.648141] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/30/2022] Open
Abstract
Sustaining and maintaining the intricate process of spermatogenesis is liable upon hormones and growth factors acting through endocrine and paracrine pathways. The Sertoli cells (SCs) are the major somatic cells present in the seminiferous tubules and are considered to be the main regulators of spermatogenesis. As each Sertoli cell supports a specific number of germ cells, thus, the final number of Sertoli cells determines the sperm production capacity. Similarly, sex hormones are also major regulators of spermatogenesis and they can determine the proliferation of Sertoli cells. In the present review, we have critically and comprehensively discussed the role of sex hormones and some other factors that are involved in Sertoli cell proliferation, differentiation and maturation. Furthermore, we have also presented a model of Sertoli cell development based upon the recent advancement in the field of reproduction. Hence, our review article provides a general overview regarding the sex hormonal pathways governing Sertoli cell proliferation and development.
Collapse
Affiliation(s)
| | - Ranjha Khan
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | | | | | | | | | - Jie Wen
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | - Xiaohua Jiang
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| |
Collapse
|