1
|
Li Y, Jiang J, Jiang R. Icariin improves erectile function in spontaneously hypertensive rats by downregulating GRK2 in penile cavernous tissue. J Sex Med 2025; 22:387-396. [PMID: 39905744 DOI: 10.1093/jsxmed/qdaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/24/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Hypertension is an independent risk factor for erectile dysfunction (ED). Icariin can improve erectile function of spontaneous hypertensive rats (SHRs). GRK2 is closely related to the phosphorylation of eNOS and endothelial function. AIM To explore whether icariin can improve erectile function in SHRs by regulating the expression of GRK2 in penile cavernous tissue. METHODS Eight-week-old WKY and SHR rats were randomly divided into four groups (n = 6 per group) as follows: WKY, WKY + icariin, SHR and SHR + icariin. The WKY + icariin and SHR + icariin groups were treated with 10 mg/kg/day icariin. After 4 weeks, the ICPmax/mean arterial pressure (MAP), serum testosterone, the levels of GRK2, p-AKT/AKT, p-eNOS/eNOS, and caspase-3; the protein interaction between GRK2 and AKT; the levels of nitric oxide (NO), superoxide dismutase (SOD), and malondialdehyde (MDA); and the level of apoptosis in rat penile cavernous tissue were measured. OUTCOME The expression of GRK2 in penile cavernous tissue of SHR was significantly higher than that in WKY rats, resulting in the inhibition of the AKT/eNOS/NO pathway, increased levels of oxidative stress and apoptosis, and the impairment of erectile function. RESULTS The ICPmax/MAP ratio in the SHR group was significantly lower than those in WKY and SHR + icariin groups (P < .01). In the SHR + icariin group, the expression levels of GRK2 and caspase-3, the interaction between GRK2 and AKT, the level of MDA and the rate of apoptosis in the penile cavernous tissue were significantly lower, and the levels of p-AKT and p-eNOS, the p-AKT/AKT and p-eNOS/eNOS ratios, and NO and SOD were significantly greater than those in the SHR group (P < .01). CLINICAL IMPLICATIONS Icariin may improve the erectile function of hypertension by downregulating GRK2 expression in penile cavernous tissue. STRENGTHS AND LIMITATIONS The specific mechanism via which icariin downregulates GRK2 needs to be further elucidated. CONCLUSION Icariin downregulates the expression of GRK2 in the penile cavernous tissue of SHRs, upregulates the AKT/eNOS/NO pathway, decreases oxidative stress and apoptosis, and ultimately improves erectile function.
Collapse
Affiliation(s)
- Yanke Li
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui Jiang
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
2
|
Ponamarczuk H, Światkowska M, Popielarski M. Androgenic Anabolic Steroids Cause Thiol Imbalance in the Vascular Endothelial Cells. FRONT BIOSCI-LANDMRK 2025; 30:26542. [PMID: 39862083 DOI: 10.31083/fbl26542] [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: 09/12/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Androgenic anabolic steroids (AASs) are synthetic drugs structurally related to testosterone, with the ability to bind to androgen receptors. Their uncontrolled use by professional and recreational sportspeople is a widespread problem. AAS abuse is correlated with severe damage to the cardiovascular system, including changes in homeostasis and coagulation disorders. AASs alter vascular function by blocking nitric oxide (NO)-mediated dilation, impairing endothelial growth and by potentiating vasoconstrictor signals. METHODS This paper demonstrated that long-term use of AASs (nandrolone and boldenone), negatively affects the basic cell functions of vascular endothelial cells. The susceptibility of endothelial cells to AASs depends on the expression of androgen receptors, although cells without androgen receptors can also be affected by high doses of AASs to a limited extent. Seven-day incubation with AASs diminishes endothelial cell proliferation and migration (determined by transwell and scratch migration assay) and monolayer formation (using transendothelial electrical resistance assay). RESULTS Disturbances in cell function were accompanied by downregulation of peroxiredoxins (PRDX1 and PRDX2), involved in maintaining the thiol-disulphide balance. In addition, AASs increased oxidation of the non-enzymatic thiol buffer, glutathione (GSH), reduced secretion of thiol oxidoreductase protein disulphide isomerase (PDI) from endothelial cells and affected the thiol pattern of PDI. CONCLUSIONS These changes may be related to a thiol-disulfide imbalance and vascular endothelium dysfunction, that are often correlated with abnormal platelet aggregation, inflammation, increased vascular permeability, and vascular smooth muscle cell proliferation-all of which are observed in athletes who abuse AASs.
Collapse
Affiliation(s)
- Halszka Ponamarczuk
- Department of Cytobiology and Proteomics, Medical University of Lodz, 92-215 Lodz, Poland
| | - Maria Światkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 92-215 Lodz, Poland
| | - Marcin Popielarski
- Department of Cytobiology and Proteomics, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
3
|
Devraj K, Kulkarni O, Liebner S. Regulation of the blood-brain barrier function by peripheral cues in health and disease. Metab Brain Dis 2024; 40:61. [PMID: 39671124 PMCID: PMC11645320 DOI: 10.1007/s11011-024-01468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/12/2024] [Indexed: 12/14/2024]
Abstract
The blood-brain barrier (BBB) is formed by microvascular endothelial cells which are ensembled with pericytes, astrocytes, microglia and neurons in the neurovascular unit (NVU) that is crucial for neuronal function. Given that the NVU and the BBB are highly dynamic and regulated structures, their integrity is continuously challenged by intrinsic and extrinsic factors. Herein, factors from peripheral organs such as gonadal and adrenal hormones may influence vascular function also in CNS endothelial cells in a sex- and age-dependent manner. The communication between the periphery and the CNS likely takes place in specific areas of the brain among which the circumventricular organs have a central position due to their neurosensory or neurosecretory function, owing to physiologically leaky blood vessels. In acute and chronic pathological conditions like liver, kidney, pulmonary disease, toxins and metabolites are generated that reach the brain via the circulation and may directly or indirectly affect BBB functionality via the activation of the immunes system. For example, chronic kidney disease (CKD) currently affects more than 840 million people worldwide and is likely to increase along with western world comorbidities of the cardio-vascular system in continuously ageing societies. Toxins leading to the uremic syndrome, may further lead to neurological complications such as cognitive impairment and uremic encephalopathy. Here we summarize the effects of hormones, toxins and inflammatory reactions on the brain vasculature, highlighting the urgent demand for mechanistically exploring the communication between the periphery and the CNS, focusing on the BBB as a last line of defense for brain protection.
Collapse
Affiliation(s)
- Kavi Devraj
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India.
| | - Onkar Kulkarni
- Metabolic Disorders and Neuroscience Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad, 500078, Telangana, India
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Partner Site Frankfurt, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
4
|
van Eeghen SA, Nokoff NJ, Vosters TG, Oosterom-Eijmael MJ, Cherney DZ, van Valkengoed IG, Choi YJ, Pyle L, Bjornstad P, den Heijer M, van Raalte DH. Unraveling Sex Differences in Kidney Health and CKD: A Review of the Effect of Sex Hormones. Clin J Am Soc Nephrol 2024; 20:01277230-990000000-00516. [PMID: 39671256 PMCID: PMC11835196 DOI: 10.2215/cjn.0000000642] [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: 08/27/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Sexual dimorphism plays an important role in the pathogenesis and progression of CKD. Men with CKD often exhibit faster kidney function decline, leading to higher rates of kidney failure and mortality compared with women. Studies suggest that sex hormones may influence this apparent dimorphism, although the mechanisms underlying these influences remain poorly understood. In this review, we first summarize recent findings on sex differences in the prevalence and progression of CKD. Subsequently, we will focus on ( 1 ) the role of sex hormones in these sex differences, ( 2 ) kidney structural and hemodynamic differences between men and women, ( 3 ) the influence of sex hormones on pathophysiological processes leading to kidney disease, including glomerular hyperfiltration and key pathways involved in kidney inflammation and fibrosis, and finally, focus on the consequences of the underrepresentation of women in clinical trials. Understanding these sex differences is critical for advancing precision medicine and improving outcomes for both men and women with CKD.
Collapse
Affiliation(s)
- Sarah A. van Eeghen
- Department of Internal Medicine, Center of Expertise on Gender Dysphoria, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Natalie J. Nokoff
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Taryn G. Vosters
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Maartina J.P. Oosterom-Eijmael
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - David Z.I. Cherney
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada
| | - Irene G.M. van Valkengoed
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura Pyle
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Petter Bjornstad
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- Division of Endocrinology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Martin den Heijer
- Department of Internal Medicine, Center of Expertise on Gender Dysphoria, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Daniël H. van Raalte
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Cardiovascular sciences Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [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: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
6
|
Yarak N, El Khoury J, Coloby P, Bart S, Abdessater M. Idiopathic recurrent ischemic priapism: a review of current literature and an algorithmic approach to evaluation and management. Basic Clin Androl 2024; 34:21. [PMID: 39627696 PMCID: PMC11616154 DOI: 10.1186/s12610-024-00237-y] [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: 07/23/2024] [Accepted: 09/06/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Stuttering priapism is characterized by recurrent, self-limited episodes of penile erection lasting from a few minutes to a maximum of three hours, often resolving spontaneously. These episodes can occur with or without sexual stimulation. If not treated promptly and effectively, stuttering priapism can severely impact a patient's quality of life, leading to significant psychological distress and anxiety related to sexual performance. Although it has been associated with various hematological disorders and pharmacological treatments, many cases of stuttering priapism remain idiopathic, meaning they have no identifiable cause. Currently, no conclusive randomized clinical trials exist on the management of idiopathic stuttering priapism. This study aims to review the existing literature on the pathophysiology and management of idiopathic stuttering priapism and propose an algorithm to assist physicians in its evaluation and treatment. RESULTS A systematic literature review was conducted using the PubMed database, focusing on the terms "idiopathic," "stuttering," "ischemic," and "priapism." The search identified 23 relevant references published between 1991 and 2022. The selection and analysis of these studies adhered to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, and results were described qualitatively. Recent research into the effectiveness, sustainability, tolerability, and side effects of various treatments for idiopathic stuttering priapism has enhanced the understanding of its underlying molecular mechanisms. Various treatments, targeting different mechanisms, have been identified that can potentially reduce the frequency and severity of episodes and improve patient outcomes. CONCLUSION Current research predominantly addresses the acute treatment of idiopathic stuttering priapism rather than strategies to alter the disease's overall course. The limited number of treatment reviews, case reports, and the low level of evidence available, combined with the absence of randomized clinical trials, prevent the establishment of a consensus on treatment protocols. As a result, idiopathic stuttering priapism remains under-recognized and under-treated. This review proposes a management framework to help clinicians access and apply the available literature effectively, minimizing the reliance on extensive case reports and review articles.
Collapse
|
7
|
Chen Z, Jiang J, Jiang R. A low testosterone level impairs erectile function by increasing endocan expression in rat penile corpus cavernosum. J Sex Med 2024; 21:663-670. [PMID: 38972662 DOI: 10.1093/jsxmed/qdae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND The mechanism by which a state of low testosterone leads to erectile dysfunction (ED) has not been determined. Endocan is a novel marker of endothelial function. However, whether endocan is involved in the regulation of erectile function under low testosterone levels remains unclear. AIM In this study we sought to determine whether a low-testosterone state inhibits erectile function by regulating endocan expression in the endothelial cells of the rat penile corpus cavernosum. METHODS Thirty-six male Sprague-Dawley rats aged 8 weeks were randomly assigned to 6 groups (n = 6 per group) as follows: (1) control, (2) castration, (3) castration + testosterone treatment (treated with 3 mg/kg testosterone propionate per 2 days), (4) control + transfection (4 weeks after castration, injected with lentiviral vector (1 × 108 transduction units/mL, 10 μL), (5) castration + transfection, or (6) castration + empty transfection. One week after the injection, we measured the maximal intracavernous pressure/mean arterial pressure (ICPmax/MAP), serum testosterone and nitric oxide (NO) levels, and the expression of endocan, phospho-endothelial NO synthase (p-eNOS), eNOS, phospho-protein kinase B (p-AKT), and AKT in the rat penile corpus cavernosum. OUTCOMES Under a low-androgen state, the expression of endocan in the rat penile corpus cavernosum was significantly increased, which inhibited the AKT/eNOS/NO signaling pathway and resulted in ED. RESULTS In the castration group, the expression of endocan in the rat penile corpus cavernosum was significantly higher than that in the control group (P < .05). Additionally, the levels of p-AKT/AKT, p-eNOS/eNOS, and NO in the rat penile corpus cavernosum and ICPmax/MAP were significantly lower in the castration group than in the control group (P < .05). In the castration + transfection group compared with the castration group there was a significant decrease in the expression of endocan (P < .05) and an increase in the ratios of p-AKT/AKT, p-eNOS/eNOS, and ICPmax/MAP (P < .05) in the rat penile corpus cavernosum. CLINICAL IMPLICATIONS Downregulating the expression of endocan in the penile corpus cavernosum may be a feasible approach for treating ED caused by hypoandrogenism. STRENGTHS AND LIMITATIONS The results of this study indicte that endocan may affect NO levels and erectile function through multiple signaling pathways, but further experiments are needed to clarify the relationship between endocan and androgens. CONCLUSION A low-testosterone state inhibits the AKT/eNOS/NO signaling pathway by increasing the expression of endocan in the rat penile corpus cavernosum and impairing erectile function in rats. Decreasing the expression of endocan in the penile corpus cavernosum can improve erectile function in rats with low testosterone levels.
Collapse
Affiliation(s)
- Zhaoguo Chen
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui Jiang
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
8
|
Naelitz BD, Khooblall PS, Parekh NV, Vij SC, Rotz SJ, Lundy SD. The effect of red blood cell disorders on male fertility and reproductive health. Nat Rev Urol 2024; 21:303-316. [PMID: 38172196 DOI: 10.1038/s41585-023-00838-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 01/05/2024]
Abstract
Male infertility is defined as a failure to conceive after 12 months of unprotected intercourse owing to suspected male reproductive factors. Non-malignant red blood cell disorders are systemic conditions that have been associated with male infertility with varying severity and strength of evidence. Hereditary haemoglobinopathies and bone marrow failure syndromes have been associated with hypothalamic-pituitary-gonadal axis dysfunction, hypogonadism, and abnormal sperm parameters. Bone marrow transplantation is a potential cure for these conditions, but exposes patients to potentially gonadotoxic chemotherapy and/or radiation that could further impair fertility. Iron imbalance might also reduce male fertility. Thus, disorders of hereditary iron overload can cause iron deposition in tissues that might result in hypogonadism and impaired spermatogenesis, whereas severe iron deficiency can propagate anaemias that decrease gonadotropin release and sperm counts. Reproductive urologists should be included in the comprehensive care of patients with red blood cell disorders, especially when gonadotoxic treatments are being considered, to ensure fertility concerns are appropriately evaluated and managed.
Collapse
Affiliation(s)
- Bryan D Naelitz
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Prajit S Khooblall
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Neel V Parekh
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Sarah C Vij
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Seth J Rotz
- Department of Paediatric Hematology and Oncology, Cleveland Clinic Children's Hospital, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Scott D Lundy
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
9
|
Abstract
Endothelial cells are important constituents of blood vessels and play a critical role in vascular homeostasis. They do not only control the exchanges between the blood and the surrounding tissues, but are also essential in regulating blood flow, modulating immune-cell trafficking and controlling vascular growth and repair. Endothelial dysfunction leads to cardiovascular diseases and is characterized by deficiency in secretion of vasodilator molecules, elevated reactive oxygen species (ROS), expression of adhesion molecules and excretion of proinflammatory cytokines. The sex hormones, estrogens, androgens and progestogens, regulate endothelial functions. Because cardiovascular disease risk increases after menopause, it is believed that female hormones, estrogens and progestogens promote endothelial cell health and function whereas androgens, the male hormones, might be detrimental. However, as illustrated in the present review, the picture might not be that simple. In addition, sex influences endothelial cell physiology independently of sex hormones but at genetic level.
Collapse
Affiliation(s)
- Jerome Robert
- University Hospital of Zurich, Institute of Clinical Chemistry, Wagistrasse 14, 8952, Schlieren, Switzerland.
| |
Collapse
|
10
|
Dou M, Liang H, Liu Y, Zhang Q, Li R, Chen S, Shi B. Based on ARASENS trial: efficacy and safety of darolutamide as an emerging option of endocrinotherapy for metastatic hormone-sensitive prostate cancer-an updated systematic review and network meta-analysis. J Cancer Res Clin Oncol 2023; 149:7017-7027. [PMID: 36856851 DOI: 10.1007/s00432-023-04658-6] [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: 12/06/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE The newly published ARASENS trial has demonstrated the clinical efficacy of darolutamide for metastatic hormone-sensitive prostate cancer (mHSPC). However, the use of darolutamide as the latest first-line androgen receptor pathway inhibitor for mHSPC has not been compared with other androgen receptor targeted agents (ARTAs). Given the lack of head-to-head randomized trials, we performed this updated meta-analysis to conduct indirect comparison for the efficacy and safety of darolutamide with other new-generation ARTAs. METHODS By searching the databases of PubMed, Scopus, Cochrane Library, and Embase, 9 large randomized controlled trials evaluating ARTAs for mHSPC patients were eventually screened according to PRISMA. We extracted data from overall survival, castration-resistant progression, and adverse events for network meta-analysis using the Bayesian and standard frequentist models. RESULTS Darolutamide combination emerged with superiority (HR = 0.68, 95%CrI = 0.57-0.81) among four androgen receptor inhibitors for patients with high Gleason score (HR = 0.71, 95%CrI = 0.59-0.86). Darolutamide was best tolerated in several androgen suppression-related adverse events (AEs). CONCLUSION Darolutamide appears to be an optional androgen receptor inhibitor for mHSPC patients, especially for patients with Gleason score ≥ 8. Its well-tolerated characteristic may provide a preferred drug option for patients with poor cardiovascular function and bone health.
Collapse
Affiliation(s)
- Maoyang Dou
- Department of Urology, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Hao Liang
- Department of Urology, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Yang Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
- Department of Urology, Weifang People's Hospital, Weifang Medical University, Weifang, Shandong, China
| | - Qiujie Zhang
- Department of Health Care, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Ruowen Li
- Department of Hernia and Abdominal Wall Surgery, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China.
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China.
| |
Collapse
|
11
|
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
|
12
|
Grandys M, Majerczak J, Frolow M, Chlopicki S, Zoladz JA. Training-induced impairment of endothelial function in track and field female athletes. Sci Rep 2023; 13:3502. [PMID: 36859449 PMCID: PMC9977863 DOI: 10.1038/s41598-023-30165-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Professional athletes are often exposed to high training loads that may lead to overfatigue, overreaching and overtraining that might have a detrimental effects on vascular health. We determined the effects of high training stress on endothelial function assessed by the flow-mediated dilation (FMD) and markers of glycocalyx shedding. Vascular examination as well as broad biochemical, hormonal and cardiometabolic evaluation of sprint and middle-distance female runners were performed after 2 months of preparatory training period and compared to age-matched control group of women. Female athletes presented with significantly reduced FMD (p < 0.01) and higher basal serum concentrations of hyaluronan (HA) and syndecan-1 (SDC-1) (p < 0.05 and p < 0.001, respectively), that was accompanied by significantly lower basal serum testosterone (T) and free testosterone (fT) concentrations (p < 0.05) and higher cortisol (C) concentration (p < 0.05). It resulted in significantly lower T/C and fT/C ratios in athletes when compared to controls (p < 0.01). Moreover, fT/C ratio were significantly positively correlated to FMD and negatively to HA concentrations in all studied women. Accordingly, the training load was significantly negatively correlated with T/C, fT/C and FMD and positively with the concentrations of HA and SDC-1. We concluded that young female track and field athletes subjected to physical training developed impairment of endothelial function that was associated with anabolic-catabolic hormone balance disturbances. Given that training-induced impairment of endothelial function may have a detrimental effects on vascular health, endothelial status should be regularly monitored in the time-course of training process to minimalize vascular health-risk in athletes.
Collapse
Affiliation(s)
- Marcin Grandys
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Ul. Skawinska 8, 31-066, Krakow, Poland.
| | - Joanna Majerczak
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Ul. Skawinska 8, 31-066, Krakow, Poland
| | - Marzena Frolow
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.,Department of Experimental Pharmacology, Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Jerzy A Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Ul. Skawinska 8, 31-066, Krakow, Poland.
| |
Collapse
|
13
|
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
|
14
|
Armeni E, Lambrinoudaki I. Menopause, androgens, and cardiovascular ageing: a narrative review. Ther Adv Endocrinol Metab 2022; 13:20420188221129946. [PMID: 36325501 PMCID: PMC9619256 DOI: 10.1177/20420188221129946] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/09/2022] [Indexed: 11/07/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide; however, women tend to be less affected than men during their reproductive years. The female cardiovascular risk increases significantly around the time of the menopausal transition. The loss of the protective action of ovarian oestrogens and the circulating androgens has been implicated in possibly inducing subclinical and overt changes in the cardiovascular system after the menopausal transition. In vitro studies performed in human or animal cell lines demonstrate an adverse effect of testosterone on endothelial cell function and nitric oxide bioavailability. Cohort studies evaluating associations between testosterone and/or dehydroepiandrosterone and subclinical vascular disease and clinical cardiovascular events show an increased risk for women with more pronounced androgenicity. However, a mediating effect of insulin resistance is possible. Data on cardiovascular implications following low-dose testosterone treatment in middle-aged women or high-dose testosterone supplementation for gender affirmatory purposes remain primarily inconsistent. It is prudent to consider the possible adverse association between testosterone and endothelial function during the decision-making process of the most appropriate treatment for a postmenopausal woman.
Collapse
Affiliation(s)
| | - Irene Lambrinoudaki
- Second Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Musicki B, Burnett AL. Testosterone Deficiency in Sickle Cell Disease: Recognition and Remediation. Front Endocrinol (Lausanne) 2022; 13:892184. [PMID: 35592776 PMCID: PMC9113536 DOI: 10.3389/fendo.2022.892184] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022] Open
Abstract
Hypogonadism is common in men with sickle cell disease (SCD) with prevalence rates as high as 25%. Testicular failure (primary hypogonadism) is established as the principal cause for this hormonal abnormality, although secondary hypogonadism and compensated hypogonadism have also been observed. The underlying mechanism for primary hypogonadism was elucidated in a mouse model of SCD, and involves increased NADPH oxidase-derived oxidative stress in the testis, which reduces protein expression of a steroidogenic acute regulatory protein and cholesterol transport to the mitochondria in Leydig cells. In all men including those with SCD, hypogonadism affects physical growth and development, cognition and mental health, sexual function, as well as fertility. However, it is not understood whether declines in physical, psychological, and social domains of health in SCD patients are related to low testosterone, or are consequences of other abnormalities of SCD. Priapism is one of only a few complications of SCD that has been studied in the context of hypogonadism. In this pathologic condition of prolonged penile erection in the absence of sexual excitement or stimulation, hypogonadism exacerbates already impaired endothelial nitric oxide synthase/cGMP/phosphodiesterase-5 molecular signaling in the penis. While exogenous testosterone alleviates priapism, it disadvantageously decreases intratesticular testosterone production. In contrast to treatment with exogenous testosterone, a novel approach is to target the mechanisms of testosterone deficiency in the SCD testis to drive endogenous testosterone production, which potentially decreases further oxidative stress and damage in the testis, and preserves sperm quality. Stimulation of translocator protein within the transduceosome of the testis of SCD mice reverses both hypogonadism and priapism, without affecting intratesticular testosterone production and consequently fertility. Ongoing research is needed to define and develop therapies that restore endogenous testosterone production in a physiologic, mechanism-specific fashion without affecting fertility in SCD men.
Collapse
|
16
|
Ferreira C, Trindade F, Ferreira R, Neves JS, Leite-Moreira A, Amado F, Santos M, Nogueira-Ferreira R. Sexual dimorphism in cardiac remodeling: the molecular mechanisms ruled by sex hormones in the heart. J Mol Med (Berl) 2021; 100:245-267. [PMID: 34811581 DOI: 10.1007/s00109-021-02169-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/16/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is growing in prevalence, due to an increase in aging and comorbidities. Heart failure with reduced ejection fraction (HFrEF) is more common in men, whereas heart failure with preserved ejection fraction (HFpEF) has a higher prevalence in women. However, the reasons for these epidemiological trends are not clear yet. Since HFpEF affects mostly postmenopausal women, sex hormones should play a pivotal role in HFpEF development. Furthermore, for HFpEF, contrary to HFrEF, effective therapeutic approaches are missing. Interestingly, studies evidenced that some therapies can have better results in women than in HFpEF men, emphasizing the necessity of understanding these observations at a molecular level. Thus, herein, we review the molecular mechanisms of estrogen and androgen actions in the heart in physiological conditions and explain how its dysregulation can lead to disease development. This clarification is essential in the road for an effective personalized management of HF, particularly HFpEF, towards the development of sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Cláudia Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Fábio Trindade
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - João Sérgio Neves
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mário Santos
- Department of Cardiology, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rita Nogueira-Ferreira
- Department of Surgery and Physiology, Cardiovascular R&D Center (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal.
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
17
|
Luberti FR, Reside TL, Bonin PL, Carré JM. Development of a single-dose intranasal testosterone administration paradigm for use in men and women. Horm Behav 2021; 136:105046. [PMID: 34488062 DOI: 10.1016/j.yhbeh.2021.105046] [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: 04/24/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022]
Abstract
For over two decades, researchers in the field of human social neuroendocrinology have been using single-dose pharmacological challenge protocols to determine the causal effects of testosterone on psychological, behavioural, and neural processes. Most of these single-dose administration studies have so far used (1) single-sex samples and (2) varying modes of testosterone administration (intramuscular, transdermal, sublingual, and intranasal) that produced vastly different dose-response curves. Moreover, whereas studies with male participants increased men's testosterone concentrations within the high normal physiological range, studies with women typically increased testosterone concentrations to supraphysiological levels. The purpose of this study was to develop a single-dose administration protocol using intranasal testosterone that would produce a proportionally similar rise in testosterone for both sexes. We found that an 11 mg intranasal testosterone dose in men and a 0.3 mg dose in women raised testosterone concentrations to the high normal physiological range for each sex, producing similar dose-response dynamics in both sexes. This paradigm will allow researchers to design studies with mixed-sex samples that test physiologically plausible sex differences/similarities in the causal effects of testosterone. It will also provide a replicable protocol to examine the possible adaptive functions of acute increases in testosterone in both sexes.
Collapse
Affiliation(s)
| | - Tracy-Lynn Reside
- Department of Psychology, Nipissing University, North Bay, ON, Canada; School of Physical Health and Education, Nipissing University, North Bay, ON, Canada
| | - Pierre L Bonin
- Family Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Justin M Carré
- Department of Psychology, Nipissing University, North Bay, ON, Canada.
| |
Collapse
|
18
|
Yazawa T, Sato T, Nemoto T, Nagata S, Imamichi Y, Kitano T, Sekiguchi T, Uwada J, Islam MS, Mikami D, Nakajima I, Takahashi S, Khan MRI, Suzuki N, Umezawa A, Ida T. 11-Ketotestosterone is a major androgen produced in porcine adrenal glands and testes. J Steroid Biochem Mol Biol 2021; 210:105847. [PMID: 33609691 DOI: 10.1016/j.jsbmb.2021.105847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Porcine steroid hormone profiles have some unique characteristics. We previously studied human and murine steroidogenesis using steroidogenic cells-derived from mesenchymal stem cells (MSCs). To investigate porcine steroidogenesis, we induced steroidogenic cells from porcine subcutaneous preadipocytes (PSPA cells), which originate from MSCs. Using cAMP, adenovirus-mediated introduction of steroidogenic factor-1 (SF-1)/adrenal 4-binding protein (Ad4BP) induced the differentiation of PSPA cells into sex steroid-producing cells. Introducing SF-1/Ad4BP also induced the aldo-keto reductase 1C1 (AKR1C1) gene. Porcine AKR1C1 had 17β-hydroxysteroid dehydrogenase activity, which converts androstenedione and 11-ketoandrostenedione into testosterone (T) and 11-ketotestosteorne (11KT). Furthermore, differentiated cells expressed hydroxysteroid 11β-dehydrogenase 2 (HSD11B2) and produced 11KT. HSD11B2 was expressed in testicular Leydig cells and the adrenal cortex. 11KT was present in the plasma of both immature male and female pigs, with slightly higher levels in the male pigs. T levels were much higher in the male pigs. It is noteworthy that in the female pigs, the 11KT levels were >10-fold higher than the T levels. However, castration altered the 11KT and T plasma profiles in the male pigs to near those of the females. 11KT induced endothelial nitric oxide synthase (eNOS) in porcine vascular endothelial cells. These results indicate that 11KT is produced in porcine adrenal glands and testes, and may regulate cardiovascular functions through eNOS expression.
Collapse
Affiliation(s)
- Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan.
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka 830-0011, Japan
| | - Takahiro Nemoto
- Department of Physiology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Sayaka Nagata
- Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshitaka Imamichi
- Department of Pharmacology, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | - Takeshi Kitano
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Toshio Sekiguchi
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| | - Junsuke Uwada
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | | | - Daisuke Mikami
- Department of Nephrology, University of Fukui, Fukui 910-1193, Japan
| | - Ikuyo Nakajima
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki 305-0901, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | - Md Rafiqul Islam Khan
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan; Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Nobuo Suzuki
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| | - Akihiro Umezawa
- Department of Reproduction, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Takanori Ida
- Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
19
|
Yan LT, Yang ZH, Lin H, Jiang J, Jiang R. Effects of androgen on extracellular vesicles from endothelial cells in rat penile corpus cavernosum. Andrology 2021; 9:1010-1017. [PMID: 33484224 DOI: 10.1111/andr.12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND The explicit mechanism of erectile dysfunction caused by low androgen status is unknown. It was reported that eNOS was expressed in extracellular vesicles (EVs). Androgen may regulate erectile function by affect the release of EVs from endothelial cells. OBJECTIVES To investigate whether androgen affects the production of EVs and nitric oxide (NO) in endothelial cells of rat penile corpus cavernosum. MATERIALS AND METHODS Endothelial cells of rat penile corpus cavernosum were isolated and purified from 6-week-old healthy male Sprague Dawley (SD) rats. Endothelial cells were treated with different concentrations of dihydrotestosterone (DHT) in a cell culture medium as follows: no-androgen group (NA group, DHT 0 nmol/L), very-low androgen group (VLA group, DHT 0.1 nmol/L), low androgen group (LA group, DHT 1 nmol/L), and physiological concentrations androgen group (PA group, DHT 10 nmol/L). After 24 h, EVs of supernatant in each group were isolated and identified. The content of EVs and NO in the supernatant and the expression of CD9, CD63, TSG101, and eNOS in EVs were detected. RESULTS Positive expression of CD9, CD63, TSG101, and eNOS was found in isolated EVs. The concentration of EVs was lower in the NA group compared with other groups (p < 0.01). The expression of eNOS and the concentration of NO was lower in the NA group than that in other groups (p < 0.05); it was lower in the VLA group than that in the LA group (p < 0.05) and lower in LA group than that in PA group (p < 0.05). When the concentration of DHT in endothelial cell culture medium ranged from 0 to 10 nmol/L, the concentration of DHT was positively correlated with the content of EVs and NO. CONCLUSION Decrease in eNOS-expressing EVs is one mechanism of NO reduction in endothelial cells of rat corpus cavernosum caused by low androgen levels.
Collapse
Affiliation(s)
- Ling-Tao Yan
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhi-Hui Yang
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haocheng Lin
- Department of Urology and Andrology, Peking University Third Hospital, Beijing, China
| | - Jun Jiang
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Jiang
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nephropathy Clinical Medical Research Center of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
20
|
Effect of Yishenjiangyafang on Plasma Metabolomics in Senile Spontaneously Hypertensive Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8868267. [PMID: 33927779 PMCID: PMC8049789 DOI: 10.1155/2021/8868267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/11/2021] [Accepted: 03/25/2021] [Indexed: 12/03/2022]
Abstract
Objectives Yishenjiangyafang is a traditional Chinese medicine used to clinically treat hypertension. This study aimed to explore the effect of yishenjiangyafang on plasma metabolomics in senile spontaneously hypertensive rats (SHRs). Methods Twelve 50-week-old SHR (6 males and 6 females) were randomly divided into two groups: a treatment group, in which rats were intragastrically administered with yishenjiangyafang (10.08 g kg−1·d−1), and a model group, in which all SHRs were administered the same volume of saline. Six age- and gender-matched Wistar–Kyoto (WKY) rats were used as the control group. Treatment was given for 6 days per week and lasted for 8 weeks. Systolic and diastolic blood pressures of the rats were measured with the noninvasive tail artery pressure measurement system. An ultraperformance liquid chromatography quadruple electrostatic field orbit (UPLC-Q-Exactive) was used to determine metabolite changes in the plasma of SHR rats before and after yishenjiangyafang treatment in the treatment group as well as in the model and control groups. Results After yishenjiangyafang treatment, SHRs had significant lower blood pressure. Using UPLC-Q-Exactive, we identified 26 metabolic targets of yishenjiangyafang in aged SHRs and revealed that yishenjiangyafang targeted four major metabolic pathways, linoleic acid metabolism, glycerophospholipid metabolism, arginine and proline metabolism, and steroid hormone biosynthesis. Conclusion Yishenjiangyafang decreases the blood pressure of SHRs at least in part through targeting of four major metabolic pathways. Our study illustrates mechanisms underlying the clinical application of yishenjiangyafang in the treatment of hypertensive patients.
Collapse
|
21
|
Chung CC, Lin YK, Kao YH, Lin SH, Chen YJ. Physiological testosterone attenuates profibrotic activities of rat cardiac fibroblasts through modulation of nitric oxide and calcium homeostasis. Endocr J 2021; 68:307-315. [PMID: 33115984 DOI: 10.1507/endocrj.ej20-0344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Testosterone deficiency is associated with poor prognosis among patients with chronic heart failure (HF). Physiological testosterone improves the exercise capacity of patients with HF. In this study, we evaluated whether treatment with physiological testosterone contributes to anti-fibrogenesis by modifying calcium homeostasis in cardiac fibroblasts and we studied the underlying mechanisms. Nitric oxide (NO) analyses, calcium (Ca2+) fluorescence, and Western blotting were performed in primary isolated rat cardiac fibroblasts with or without (control cells) testosterone (10, 100, 1,000 nmol/L) treatment for 48 hours. Physiological testosterone (10 nmol/L) increased NO production and phosphorylation at the inhibitory site of the inositol trisphosphate (IP3) receptor, thereby reducing Ca2+ entry, phosphorylated Ca2+/calmodulin-dependent protein kinase II (CaMKII) expression, type I and type III pro-collagen production. Non-physiological testosterone-treated fibroblasts exhibited similar NO and collagen production capabilities as compared to control (testosterone deficient) fibroblasts. These effects were blocked by co-treatment with NO inhibitor (L-NG-nitro arginine methyl ester [L-NAME], 100 μmol/L). In the presence of the IP3 receptor inhibitor (2-aminoethyl diphenylborinate [2-APB], 50 μmol/L), testosterone-deficient and physiological testosterone-treated fibroblasts exhibited similar phosphorylated CaMKII expression. When treated with 2-APB or CaMKII inhibitor (KN93, 10 μmol/L), testosterone-deficient and physiological testosterone-treated fibroblasts exhibited similar type I, and type III collagen production. In conclusion, physiological testosterone activates NO production, and attenuates the IP3 receptor/Ca2+ entry/CaMKII signaling pathway, thereby inhibiting the collagen production capability of cardiac fibroblasts.
Collapse
Affiliation(s)
- Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shyh-Hsiang Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
22
|
Joice GA, Liu JL, Burnett AL. Medical treatment of recurrent ischaemic priapism: a review of current molecular therapeutics and a new clinical management paradigm. BJU Int 2021; 127:498-506. [PMID: 33606327 DOI: 10.1111/bju.15370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES To examine the current molecular therapeutics in the medical treatment of recurrent ischemic priapism (RIP). To propose a stepwise clinical management paradigm for the treatment of RIP. METHODS We performed a literature search using the PubMed database for the terms 'recurrent ischemic priapism' and 'stuttering priapism' up until December 2020. We assessed pre-clinical and clinical studies regarding medical management of RIP and molecular pathophysiology. Case series and randomized trials were evaluated by study quality and patient outcomes to determine a potential clinical management scheme. RESULTS Recent research has fostered an improved understanding of the underlying molecular pathophysiology of RIP that has paved the way forward for developing new therapeutic agents. Medications targeting neurovascular, hormonal and haematological mechanisms associated with RIP show great promise towards remedying this condition. A host of therapeutic agents operating across different mechanistic directions may be implemented according to a clinical management scheme to potentially optimize RIP outcomes. CONCLUSION RIP remains a medically neglected condition with current management focused on treating the acute condition rather than modulating the course of disease. Continued research into the molecular mechanisms of RIP and standardized clinical pathways can improve the quality of care for patients suffering from this condition.
Collapse
Affiliation(s)
- Gregory A Joice
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James L Liu
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arthur L Burnett
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Jeon YK, Shin MJ, Saini SK, Custodero C, Aggarwal M, Anton SD, Leeuwenburgh C, Mankowski RT. Vascular dysfunction as a potential culprit of sarcopenia. Exp Gerontol 2020; 145:111220. [PMID: 33373710 DOI: 10.1016/j.exger.2020.111220] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/08/2023]
Abstract
Aging-related changes to biological structures such as cardiovascular and musculoskeletal systems contribute to the development of comorbid conditions including cardiovascular disease and frailty, and ultimately lead to premature death. Although, frail older adults often demonstrate both cardiovascular and musculoskeletal comorbidities, the etiology of sarcopenia, and especially the contribution of cardiovascular aging is unclear. Aging-related vascular calcification is prevalent in older adults and is a known risk factor for cardiovascular disease and death. The effect vascular calcification has on function during aging is not well understood. Emerging findings suggest vascular calcification can impact skeletal muscle perfusion, negatively affecting nutrient and oxygen delivery to skeletal muscle, ultimately accelerating muscle loss and functional decline. The present review summarizes existing evidence on the biological mechanisms linking vascular calcification with sarcopenia during aging.
Collapse
Affiliation(s)
- Yun Kyung Jeon
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA; Division of Endocrinology and Metabolism, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Myung Jun Shin
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA; Department of Rehabilitation Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Sunil Kumar Saini
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | - Carlo Custodero
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA; Dipartimento Interdisciplinare di Medicina, Clinica Medica Cesare Frugoni, University of Bari Aldo Moro, Bari, Italy
| | - Monica Aggarwal
- Department of Medicine, Division of Cardiovascular Medicine, University of Florida, FL, USA
| | - Stephen D Anton
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | | | - Robert T Mankowski
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
24
|
Gur S, Alzweri L, Yilmaz‐Oral D, Kaya‐Sezginer E, Abdel‐Mageed AB, Dick B, Sikka SC, Volkan Oztekin C, Hellstrom WJG. Testosterone positively regulates functional responses and nitric oxide expression in the isolated human corpus cavernosum. Andrology 2020; 8:1824-1833. [DOI: 10.1111/andr.12866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/30/2020] [Accepted: 07/08/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Serap Gur
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
- Department of Pharmacology Faculty of Pharmacy Ankara University Ankara Turkey
| | - Laith Alzweri
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
- Division of Urology, Department of Surgery University of Texas Medical Branch Galveston TX USA
| | - Didem Yilmaz‐Oral
- Department of Pharmacology Faculty of Pharmacy Cukurova University Adana Turkey
| | - Ecem Kaya‐Sezginer
- Department of Biochemistry Faculty of Pharmacy Ankara University Ankara Turkey
| | - Asim B Abdel‐Mageed
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Brian Dick
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Suresh C. Sikka
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Cetin Volkan Oztekin
- Department of Urology Faculty of Medicine University of KyreniaTurkish Republic of North Cyprus Girne, Mersin 10 Turkey
| | | |
Collapse
|
25
|
Sesti F, Pofi R, Minnetti M, Tenuta M, Gianfrilli D, Isidori AM. Late-onset hypogonadism: Reductio ad absurdum of the cardiovascular risk-benefit of testosterone replacement therapy. Andrology 2020; 8:1614-1627. [PMID: 32737921 DOI: 10.1111/andr.12876] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Low testosterone (T) level is considered a marker of poor cardiovascular health. Ten years ago, the Testosterone in Older Men with Mobility Limitations (TOM) trial was discontinued due to a higher number of adverse events in men receiving T compared with placebo. Since then, several studies have investigated the risks of T replacement therapy (TRT) in late-onset hypogonadism (LOH). OBJECTIVE To review the mechanism by which TRT could damage the cardiovascular system. MATERIALS AND METHODS Comprehensive literature search of recent clinical and experimental studies. RESULTS The mechanisms of T-mediated coronary vasodilation were reviewed with emphasis on calcium-activated and ATP-sensitive potassium ion channels. We showed how T regulates endothelial nitric oxide synthase (eNOS) and phosphoinositide 3-kinase/protein kinase B/eNOS signaling pathways in vessel walls and its direct effects on cardiomyocytes via β1-adrenergic and ryanodine receptors and provided data on myocardial infarction and heart failure. Vascular smooth muscle senescence could be explained by the modulation of growth factors, matrix metalloproteinase-2, and angiotensin II by T. Furthermore, leukocyte trafficking, facilitated by changes in TNF-α, could explain some of the effects of T on atheromatous plaques. Conflicting data on prothrombotic risk linked to platelet aggregation inhibition via NO-triggered arachidonate synthesis or increased aggregability due to enhanced thromboxane A in human platelets provide evidence regarding the hypotheses on plaque maturation and rupture risk. The effects of T on cardiac electrophysiology and oxygen delivery were also reviewed. DISCUSSION The effects of TRT on the cardiovascular system are complex. Although molecular studies suggest a potential benefit, several clinical observations reveal neutral or occasionally detrimental effects, mostly due to confounding factors. CONCLUSIONS Attempts to demonstrate that TRT damages the cardiovascular system via systematic analysis of the putative mechanisms led to the contradiction of the initial hypothesis. Current evidence indicates that TRT is safe once other comorbidities are addressed.
Collapse
Affiliation(s)
- Franz Sesti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Minnetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Marta Tenuta
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
26
|
Garcia V, Park EJ, Siragusa M, Frohlich F, Mahfuzul Haque M, Pascale JV, Heberlein KR, Isakson BE, Stuehr DJ, Sessa WC. Unbiased proteomics identifies plasminogen activator inhibitor-1 as a negative regulator of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:9497-9507. [PMID: 32300005 PMCID: PMC7196906 DOI: 10.1073/pnas.1918761117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.
Collapse
Affiliation(s)
- Victor Garcia
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Mauro Siragusa
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Florian Frohlich
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
- Department of Biology/Chemistry, Molecular Membrane Biology Section, University of Osnabrück, 49076 Osnabrück, Germany
| | - Mohammad Mahfuzul Haque
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Katherine R Heberlein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|
27
|
Musicki B, Burnett AL. Mechanisms underlying priapism in sickle cell disease: targeting and key innovations on the preclinical landscape. Expert Opin Ther Targets 2020; 24:439-450. [PMID: 32191546 DOI: 10.1080/14728222.2020.1745188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Priapism is prolonged penile erection in the absence of sexual arousal or desire and is a devastating condition affecting millions of patients with sickle cell disease (SCD) globally. Available drug treatments for SCD-related priapism remain limited and have been primarily reactive rather than preventive. Hence, there is an unmet need for new drug targets and pharmacologic therapies.Areas covered: We examine the molecular mechanisms underlying SCD-associated priapism evaluated mostly in animal models. In mouse models of SCD, molecular defects of priapism operating at the cavernous tissue level include reduced tonic NO/cGMP signaling, elevated oxidative/nitrosative stress, vascular adhesion molecule derangements, excessive adenosine and opiorphin signaling, dysregulated vasoconstrictive RhoA/ROCK signaling, and testosterone deficiency. We discuss the consequences of downregulated cGMP-dependent phosphodiesterase type 5 (PDE5) activity in response to these molecular signaling derangements, as the main effector mechanism causing unrestrained cavernous tissue relaxation that results in priapism.Expert opinion: Basic science studies are crucial for understanding the underlying pathophysiology of SCD-associated priapism. Understanding the molecular mechanisms could unearth new therapeutic targets for this condition based on these mechanisms. Treatment options should aim to improve deranged erection physiology regulatory signaling to prevent priapism and potentially restore or preserve erectile function.
Collapse
Affiliation(s)
- Biljana Musicki
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arthur L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Lorigo M, Mariana M, Lemos MC, Cairrao E. Vascular mechanisms of testosterone: The non-genomic point of view. J Steroid Biochem Mol Biol 2020; 196:105496. [PMID: 31655180 DOI: 10.1016/j.jsbmb.2019.105496] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/26/2019] [Accepted: 10/07/2019] [Indexed: 01/19/2023]
Abstract
Testosterone (T) is the predominant endogenous androgen in the bloodstream. At the vascular level, T presents genomic and non-genomic effects, and both effects may overlap. The genomic actions assume that androgens can freely cross the plasma membrane of target cells and bind to nuclear androgen receptors, inducing gene transcription and protein synthesis. The non-genomic effects have a more rapid onset and may be related to the interaction with protein/receptor/ion channels of the plasma membrane. The key T effect at the vascular level is vasorelaxation, which is primarily due to its rapid effect. Thus, the main purpose of this review is to discuss the T non-genomic effects at the vascular level and the molecular pathways involved in its vasodilator effect observed in in vivo and in vitro studies. In this sense, the nuclear receptor activation, the influence of vascular endothelium and the activation or inhibition of ion channels (potassium and calcium channels, respectively) will be reviewed regarding all the data that corroborated or not. Moreover, this review also provides a brief update on the association of T with the risk factors for cardiovascular diseases, namely metabolic syndrome, type 2 diabetes mellitus, obesity, atherosclerosis, dyslipidaemia, and hypertension. In summary, in this paper we consider the non-genomic vascular mode of action of androgen in physiological conditions and the main risk factors for cardiovascular diseases.
Collapse
Affiliation(s)
- Margarida Lorigo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Melissa Mariana
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Manuel C Lemos
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Elisa Cairrao
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
29
|
Hester J, Ventetuolo C, Lahm T. Sex, Gender, and Sex Hormones in Pulmonary Hypertension and Right Ventricular Failure. Compr Physiol 2019; 10:125-170. [PMID: 31853950 DOI: 10.1002/cphy.c190011] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) encompasses a syndrome of diseases that are characterized by elevated pulmonary artery pressure and pulmonary vascular remodeling and that frequently lead to right ventricular (RV) failure and death. Several types of PH exhibit sexually dimorphic features in disease penetrance, presentation, and progression. Most sexually dimorphic features in PH have been described in pulmonary arterial hypertension (PAH), a devastating and progressive pulmonary vasculopathy with a 3-year survival rate <60%. While patient registries show that women are more susceptible to development of PAH, female PAH patients display better RV function and increased survival compared to their male counterparts, a phenomenon referred to as the "estrogen paradox" or "estrogen puzzle" of PAH. Recent advances in the field have demonstrated that multiple sex hormones, receptors, and metabolites play a role in the estrogen puzzle and that the effects of hormone signaling may be time and compartment specific. While the underlying physiological mechanisms are complex, unraveling the estrogen puzzle may reveal novel therapeutic strategies to treat and reverse the effects of PAH/PH. In this article, we (i) review PH classification and pathophysiology; (ii) discuss sex/gender differences observed in patients and animal models; (iii) review sex hormone synthesis and metabolism; (iv) review in detail the scientific literature of sex hormone signaling in PAH/PH, particularly estrogen-, testosterone-, progesterone-, and dehydroepiandrosterone (DHEA)-mediated effects in the pulmonary vasculature and RV; (v) discuss hormone-independent variables contributing to sexually dimorphic disease presentation; and (vi) identify knowledge gaps and pathways forward. © 2020 American Physiological Society. Compr Physiol 10:125-170, 2020.
Collapse
Affiliation(s)
- James Hester
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Corey Ventetuolo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Xu X, Yan Q, Liu X, Li P, Li X, Chen Y, Simoncini T, Liu J, Zhu D, Fu X. 17β-Estradiol nongenomically induces vascular endothelial H 2S release by promoting phosphorylation of cystathionine γ-lyase. J Biol Chem 2019; 294:15577-15592. [PMID: 31439665 DOI: 10.1074/jbc.ra119.008597] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Estrogen exerts its cardiovascular protective role at least in part by regulating endothelial hydrogen sulfide (H2S) release, but the underlying mechanisms remain to be fully elucidated. Estrogen exerts genomic effects, i.e. those involving direct binding of the estrogen receptor (ER) to gene promoters in the nucleus, and nongenomic effects, mediated by interactions of the ER with other proteins. Here, using human umbilical vein endothelial cells (HUVECs), immunological detection, MS-based analyses, and cGMP and H2S assays, we show that 17β-estradiol (E2) rapidly enhances endothelial H2S release in a nongenomic manner. We found that E2 induces phosphorylation of cystathionine γ-lyase (CSE), the key enzyme in vascular endothelial H2S generation. Mechanistically, E2 enhanced the interaction of membrane ERα with the Gα subunit Gαi-2/3, which then transactivated particulate guanylate cyclase-A (pGC-A) to produce cGMP, thereby activating protein kinase G type I (PKG-I). We also found that PKG-Iβ, but not PKG-Iα, interacts with CSE, leading to its phosphorylation, and rapidly induces endothelial H2S release. Furthermore, we report that silencing of either CSE or pGC-A in mice attenuates E2-induced aorta vasodilation. These results provide detailed mechanistic insights into estrogen's nongenomic effects on vascular endothelial H2S release and advance our current understanding of the protective activities of estrogen in the cardiovascular system.
Collapse
Affiliation(s)
- Xingyan Xu
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Qing Yan
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaoyun Liu
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ping Li
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaosa Li
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yiwen Chen
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa 56100, Italy
| | - Junxiu Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Dongxing Zhu
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China .,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaodong Fu
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China .,State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
31
|
Chistiakov DA, Myasoedova VA, Melnichenko AA, Grechko AV, Orekhov AN. Role of androgens in cardiovascular pathology. Vasc Health Risk Manag 2018; 14:283-290. [PMID: 30410343 PMCID: PMC6198881 DOI: 10.2147/vhrm.s173259] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular effects of android hormones in normal and pathological conditions can lead to either positive or negative effects. The reason for this variation is unknown, but may be influenced by gender-specific effects of androids, heterogeneity of the vascular endothelium, differential expression of the androgen receptor in endothelial cells (ECs) and route of androgen administration. Generally, androgenic hormones are beneficial for ECs because these hormones induce nitric oxide production, proliferation, motility, and growth of ECs and inhibit inflammatory activation and induction of procoagulant, and adhesive properties in ECs. This indeed prevents endothelial dysfunction, an essential initial step in the development of vascular pathologies, including atherosclerosis. However, androgens can also activate endothelial production of some vasoconstrictors, which can have detrimental effects on the vascular endothelium. Androgens also activate proliferation, migration, and recruitment of endothelial progenitor cells (EPCs), thereby contributing to vascular repair and restoration of the endothelial layer. In this paper, we consider effects of androgen hormones on EC and EPC function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Neurochemistry, Division of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center of Psychiatry and Narcology, Moscow, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia,
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia,
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia, .,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia,
| |
Collapse
|
32
|
Nitric oxide and the biology of pregnancy. Vascul Pharmacol 2018; 110:71-74. [PMID: 30076925 DOI: 10.1016/j.vph.2018.07.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/29/2018] [Indexed: 01/16/2023]
Abstract
Nitric oxide (NO) is a key regulator of both maternal and fetal homeostasis during pregnancy, facilitating the maternal cardio-vascular changes, fetal development and growth and adaptation to extrauterine life. Dysfunction of the NO system during pregnancy is associated to placental and vascular-related diseases such as hypertensive disorders of pregnancy (HDP) and intrauterine growth restriction (IUGR). Emerging therapeutic strategies involving NO precursors, NO donors, natural derivatives or pharmacological modulators of the NO system seem hold promise for the treatment of such conditions of pregnancy.
Collapse
|
33
|
Effect of Low Androgen Status on the Expression of P2Y Receptors in the Corpus Cavernosum of Rats. Urology 2018; 116:229.e1-229.e6. [DOI: 10.1016/j.urology.2018.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/23/2018] [Accepted: 03/14/2018] [Indexed: 01/11/2023]
|
34
|
Vanetti C, Bifari F, Vicentini LM, Cattaneo MG. Fatty acids rather than hormones restore in vitro angiogenesis in human male and female endothelial cells cultured in charcoal-stripped serum. PLoS One 2017; 12:e0189528. [PMID: 29232396 PMCID: PMC5726635 DOI: 10.1371/journal.pone.0189528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/27/2017] [Indexed: 01/18/2023] Open
Abstract
Charcoal-stripped serum (CSS) is a well-accepted method to model effects of sex hormones in cell cultures. We have recently shown that human endothelial cells (ECs) fail to growth and to undergo in vitro angiogenesis when cultured in CSS. However, the mechanism(s) underlying the CSS-induced impairment of in vitro EC properties are still unknown. In addition, whether there is any sexual dimorphism in the CSS-induced EC phenotype remains to be determined. Here, by independently studying human male and female ECs, we found that CSS inhibited both male and female EC growth and in vitro angiogenesis, with a more pronounced effect on male EC sprouting. Reconstitution of CSS with 17-β estradiol, dihydrotestosterone, or the lipophilic thyroid hormone did not restore EC functions in both sexes. On the contrary, supplementation with palmitic acid or the acetyl-CoA precursor acetate significantly rescued the CSS-induced inhibition of growth and sprouting in both male and female ECs. We can conclude that the loss of metabolic precursors (e.g., fatty acids) rather than of hormones is involved in the impairment of in vitro proliferative and angiogenic properties of male and female ECs cultured with CSS.
Collapse
Affiliation(s)
- Claudia Vanetti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Lucia M. Vicentini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
- * E-mail:
| |
Collapse
|
35
|
Musicki B, Karakus S, Akakpo W, Silva FH, Liu J, Chen H, Zirkin BR, Burnett AL. Testosterone replacement in transgenic sickle cell mice controls priapic activity and upregulates PDE5 expression and eNOS activity in the penis. Andrology 2017; 6:184-191. [PMID: 29145710 DOI: 10.1111/andr.12442] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/30/2017] [Accepted: 10/04/2017] [Indexed: 01/02/2023]
Abstract
Sickle cell disease (SCD)-associated priapism is characterized by decreased nitric oxide (NO) signaling and downregulated phosphodiesterase (PDE)5 protein expression and activity in the penis. Priapism is also associated with testosterone deficiency, but molecular mechanisms underlying testosterone effects in the penis in SCD are not known. Given the critical role of androgens in erection physiology and NO synthase (NOS)/PDE5 expression, we hypothesized that testosterone replacement to eugonadal testosterone levels reduces priapism by reversing impaired endothelial (e)NOS activity and molecular abnormalities involving PDE5. Adult male transgenic Berkeley sickle cell (Sickle) and wild-type (WT) mice were implanted with testosterone pellets, which release 1.2 μg testosterone/day for 21 days, or vehicle. After 21 days, animals underwent erectile function assessment followed by collection of blood for serum testosterone measurements, penes for molecular analysis, and seminal vesicles as testosterone-responsive tissue. Serum testosterone levels were measured by radioimmunoassay; protein expressions of PDE5, α-smooth muscle actin, eNOS and nNOS, and phosphorylation of PDE5 at Ser-92, eNOS at Ser-1177, neuronal (n) NOS at Ser-1412, and Akt at Ser-473 were measured by Western blot in penile tissue. Testosterone treatment reversed downregulated serum testosterone levels and increased (p < 0.05) the weight of seminal vesicles in Sickle mice to levels comparable to that of WT mice, indicating restored testosterone levels in Sickle mice. Testosterone treatment reduced (p < 0.05) prolonged detumescence in Sickle mice and normalized downregulated P-PDE5 (Ser-92), PDE5, P-eNOS (Ser-1177), and P-Akt (Ser-473) protein expressions in the Sickle mouse penis. Testosterone treatment did not affect P-nNOS (Ser-1412), eNOS, nNOS, or α-smooth muscle actin protein expressions in the Sickle mouse penis. In conclusion, in the mouse model of human SCD, increasing testosterone to eugonadal levels reduced priapic activity and reversed impaired Akt/eNOS activity and PDE5 protein expression in the penis.
Collapse
Affiliation(s)
- B Musicki
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - S Karakus
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - W Akakpo
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - F H Silva
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J Liu
- Department of Biochemistry and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Chen
- Department of Biochemistry and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B R Zirkin
- Department of Biochemistry and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Traish AM. Benefits and Health Implications of Testosterone Therapy in Men With Testosterone Deficiency. Sex Med Rev 2017; 6:86-105. [PMID: 29128268 DOI: 10.1016/j.sxmr.2017.10.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Testosterone (T) deficiency (TD; hypogonadism) has deleterious effects on men's health; negatively affects glycometabolic and cardiometabolic functions, body composition, and bone mineral density; contributes to anemia and sexual dysfunction; and lowers quality of life. T therapy (TTh) has been used for the past 8 decades to treat TD, with positive effects on signs and symptoms of TD. AIM To summarize the health benefits of TTh in men with TD. METHODS A comprehensive literature search was carried out using PubMed, articles relevant to TTh were accessed and evaluated, and a comprehensive summary was synthesized. MAIN OUTCOME MEASURES Improvements in signs and symptoms of TD reported in observational studies, registries, clinical trials, and meta-analyses were reviewed and summarized. RESULTS A large body of evidence provides significant valuable information pertaining to the therapeutic value of TTh in men with TD. TTh in men with TD provides real health benefits for bone mineral density, anemia, sexual function, glycometabolic and cardiometabolic function, and improvements in body composition, anthropometric parameters, and quality of life. CONCLUSION TTh in the physiologic range for men with TD is a safe and effective therapeutic modality and imparts great benefits on men's health and quality of life. Traish AM. Benefits and Health Implications of Testosterone Therapy in Men With Testosterone Deficiency. Sex Med Rev 2018;6:86-105.
Collapse
Affiliation(s)
- Abdulmaged M Traish
- Department of Urology, Boston University School of Medicine, Boson, MA, USA.
| |
Collapse
|
37
|
Xu X, Wang B, Ren C, Hu J, Greenberg DA, Chen T, Xie L, Jin K. Recent Progress in Vascular Aging: Mechanisms and Its Role in Age-related Diseases. Aging Dis 2017; 8:486-505. [PMID: 28840062 PMCID: PMC5524810 DOI: 10.14336/ad.2017.0507] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/07/2017] [Indexed: 01/13/2023] Open
Abstract
As with many age-related diseases including vascular dysfunction, age is considered an independent and crucial risk factor. Complicated alterations of structure and function in the vasculature are linked with aging hence, understanding the underlying mechanisms of age-induced vascular pathophysiological changes holds possibilities for developing clinical diagnostic methods and new therapeutic strategies. Here, we discuss the underlying molecular mediators that could be involved in vascular aging, e.g., the renin-angiotensin system and pro-inflammatory factors, metalloproteinases, calpain-1, monocyte chemoattractant protein-1 (MCP-1) and TGFβ-1 as well as the potential roles of testosterone and estrogen. We then relate all of these to clinical manifestations such as vascular dementia and stroke in addition to reviewing the existing clinical measurements and potential interventions for age-related vascular dysfunction.
Collapse
Affiliation(s)
- Xianglai Xu
- 1Zhongshan Hospital, Fudan University, Shanghai 200032, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Brian Wang
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Changhong Ren
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.,4Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University. Beijing, China
| | - Jiangnan Hu
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | | | - Tianxiang Chen
- 6Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Liping Xie
- 3Department of Urology, the First Affiliated Hospital, Zhejiang University, Zhejiang Province, China
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| |
Collapse
|
38
|
Swerdloff RS, Dudley RE, Page ST, Wang C, Salameh WA. Dihydrotestosterone: Biochemistry, Physiology, and Clinical Implications of Elevated Blood Levels. Endocr Rev 2017; 38:220-254. [PMID: 28472278 PMCID: PMC6459338 DOI: 10.1210/er.2016-1067] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/20/2017] [Indexed: 02/07/2023]
Abstract
Benefits associated with lowered serum DHT levels after 5α-reductase inhibitor (5AR-I) therapy in men have contributed to a misconception that circulating DHT levels are an important stimulus for androgenic action in target tissues (e.g., prostate). Yet evidence from clinical studies indicates that intracellular concentrations of androgens (particularly in androgen-sensitive tissues) are essentially independent of circulating levels. To assess the clinical significance of modest elevations in serum DHT and the DHT/testosterone (T) ratio observed in response to common T replacement therapy, a comprehensive review of the published literature was performed to identify relevant data. Although the primary focus of this review is about DHT in men, we also provide a brief overview of DHT in women. The available published data are limited by the lack of large, well-controlled studies of long duration that are sufficiently powered to expose subtle safety signals. Nonetheless, the preponderance of available clinical data indicates that modest elevations in circulating levels of DHT in response to androgen therapy should not be of concern in clinical practice. Elevated DHT has not been associated with increased risk of prostate disease (e.g., cancer or benign hyperplasia) nor does it appear to have any systemic effects on cardiovascular disease safety parameters (including increased risk of polycythemia) beyond those commonly observed with available T preparations. Well-controlled, long-term studies of transdermal DHT preparations have failed to identify safety signals unique to markedly elevated circulating DHT concentrations or signals materially different from T.
Collapse
Affiliation(s)
- Ronald S Swerdloff
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
| | | | - Stephanie T Page
- Division of Metabolism, Endocrinology, and Nutrition, University of Washington School of Medicine, Seattle, Washington 98195
| | - Christina Wang
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
- UCLA Clinical and Translational Science Institute, Harbor-UCLA Medical Center, and Los Angeles Biomedical Research Institute, David Geffen School of Medicine at UCLA, Torrance, California 90509
| | - Wael A Salameh
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine at UCLA, Torrance, California 90502
| |
Collapse
|
39
|
Ruamyod K, Watanapa WB, Shayakul C. Testosterone rapidly increases Ca 2+-activated K + currents causing hyperpolarization in human coronary artery endothelial cells. J Steroid Biochem Mol Biol 2017; 168:118-126. [PMID: 28223151 DOI: 10.1016/j.jsbmb.2017.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022]
Abstract
Testosterone has endothelium-dependent vasodilatory effects on the coronary artery, with some reports suggesting endothelial ion channel involvement. This study employed the whole-cell patch clamp technique to investigate the effect of testosterone on ion channels in human coronary artery endothelial cells (HCAECs) and the mechanisms involved. We found that 0.03-3μM testosterone significantly induced a rapid, concentration-dependent increase in total HCAEC current (EC50, 71.96±1.66nM; maximum increase, 59.13±8.37%; mean±SEM). The testosterone-enhanced currents consisted of small- and large-conductance Ca2+-activated K+ currents (SKCa and BKCa currents), but not Cl- and nonselective cation currents. Either a non-permeant testosterone conjugate or the non-aromatizable androgen dihydrotestosterone (DHT) could increase HCAEC currents as well. The androgen receptor antagonist flutamide prevented this testosterone, testosterone conjugate, and DHT effect, while the estrogen receptor antagonist fulvestrant did not. Incubating HCAECs with pertussis toxin or protein kinase A inhibitor H-89 largely inhibited the testosterone effect, while pre-incubation with phospholipase C inhibitor U-73122, prostacyclin inhibitor indomethacin, nitric oxide synthase inhibitor L-NAME or cytochrome P450 inhibitor MS-PPOH, did not. Finally, testosterone application induced HCAEC hyperpolarization within minutes; this effect was prevented by SKCa and BKCa current inhibitors apamin and iberiotoxin. This is the first electrophysiological demonstration of androgen-induced KCa current increase, leading to hyperpolarization, in any endothelial cell, and the first report of SKCa as a testosterone target. Our data show that testosterone rapidly increased whole-cell HCAEC SKCa and BKCa currents via a surface androgen receptor, Gi/o protein, and protein kinase A. This mechanism may explain rapid testosterone-induced coronary vasodilation seen in vivo.
Collapse
Affiliation(s)
- Katesirin Ruamyod
- Department of Physiology Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Wattana B Watanapa
- Department of Physiology Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Chairat Shayakul
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
40
|
Chenu C, Adlanmerini M, Boudou F, Chantalat E, Guihot AL, Toutain C, Raymond-Letron I, Vicendo P, Gadeau AP, Henrion D, Arnal JF, Lenfant F. Testosterone Prevents Cutaneous Ischemia and Necrosis in Males Through Complementary Estrogenic and Androgenic Actions. Arterioscler Thromb Vasc Biol 2017; 37:909-919. [PMID: 28360090 DOI: 10.1161/atvbaha.117.309219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/13/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Chronic nonhealing wounds are a substantial medical concern and are associated with morbidity and mortality; thus, new treatment strategies are required. The first step toward personalized/precision medicine in this field is probably in taking sex differences into account. Impaired wound healing is augmented by ischemia, and we previously demonstrated that 17β-estradiol exerts a major preventive effect against ischemia-induced skin flap necrosis in female mice. However, the equivalent effects of testosterone in male mice have not yet been reported. We then investigated the role of steroid hormones in male mice using a skin flap ischemia model. APPROACH AND RESULTS Castrated male mice developed skin necrosis after ischemia, whereas intact or castrated males treated with testosterone were equally protected. Testosterone can (1) activate the estrogen receptor after its aromatization into 17β-estradiol or (2) be reduced into dihydrotestosterone, a nonaromatizable androgen that activates the androgen receptor. We found that dihydrotestosterone protected castrated wild-type mice by promoting skin revascularization, probably through a direct action on resistance arteries, as evidenced using a complementary model of flow-mediated outward remodeling. 17β-estradiol treatment of castrated male mice also strongly protected them from ischemic necrosis through the activation of estrogen receptor-α by increasing skin revascularization and skin survival. Remarkably, 17β-estradiol improved skin survival with a greater efficiency than dihydrotestosterone. CONCLUSIONS Testosterone provides males with a strong protection against cutaneous necrosis and acts through both its estrogenic and androgenic derivatives, which have complementary effects on skin survival and revascularization.
Collapse
Affiliation(s)
- Caroline Chenu
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Marine Adlanmerini
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Frederic Boudou
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Elodie Chantalat
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Anne-Laure Guihot
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Céline Toutain
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Isabelle Raymond-Letron
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Patricia Vicendo
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Alain-Pierre Gadeau
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Daniel Henrion
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Jean-François Arnal
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.)
| | - Françoise Lenfant
- From the INSERM U1048, Institut de Médecine Moléculaire de Rangueil, CHU Toulouse, Université Toulouse III Paul-Sabatier, France (C.C., M.A., F.B., E.C., C.T., J.-F.A., F.L.); Département d'Anatomie-Pathologique, Ecole Nationale Vétérinaire de Toulouse, France (I.R.-L.); Laboratoire des IMRCP, UMR 5623, Université de Toulouse, Université Paul Sabatier, France (P.V.); INSERM U1034, Université de Bordeaux, Pessac, France (A.-P.G.); and MITOVASC, CARFI, INSERM U1083 and CNRS UMR6214, Université d'Angers, France (A.-L.G., D.H.).
| |
Collapse
|
41
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 458] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
42
|
Ghebre YT, Yakubov E, Wong WT, Krishnamurthy P, Sayed N, Sikora AG, Bonnen MD. Vascular Aging: Implications for Cardiovascular Disease and Therapy. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2016; 6:183. [PMID: 28932625 PMCID: PMC5602592 DOI: 10.4172/2161-1025.1000183] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence and prevalence of cardiovascular disease is highest among the elderly, in part, due to deleterious effects of advancing age on the heart and blood vessels. Aging, a known cardiovascular risk factor, is progressively associated with structural and functional changes to the vasculature including hemodynamic disturbance due to increased oxidative stress, premature cellular senescence and impairments in synthesis and/or secretion of endothelium-derived vasoactive molecules. These molecular and physiological changes lead to vessel wall stiffening and thickening, as well as other vascular complications that culminate to loss of vascular tone regulation and endothelial function. Intriguingly, the vessel wall, a biochemically active structure composed of collagen, connective tissue, smooth muscle and endothelial cells, is adversely affected by processes involved in premature or normal aging. Notably, the inner most layer of the vessel wall, the endothelium, becomes senescent and dysfunctional with advancing age. As a result, its ability to release vasoactive molecules such as acetylcholine (ACh), prostacyclin (PGI2), endothelium-derived hyperpolarizing factor (EDHF), and nitric oxide (NO) is reduced and the cellular response to these molecules is also impaired. By contrast, the vascular endothelium increases its generation and release of reactive oxygen (ROS) and nitrogen (RNS) species, vasoconstrictors such as endothelin (ET) and angiotensin (AT), and endogenous inhibitors of NO synthases (NOSs) to block NO. This skews the balance of the endothelium in favor of the release of highly tissue reactive and harmful molecules that promote DNA damage, telomere erosion, senescence, as well as stiffened and hardened vessel wall that is prone to the development of hypertension, diabetes, atherosclerosis and other cardiovascular risk factors. This Review discusses the impact of advancing age on cardiovascular health, and highlights the cellular and molecular mechanisms that underlie age-associated vascular changes. In addition, the role of pharmacological interventions in preventing or delaying age-related cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Yohannes T Ghebre
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Eduard Yakubov
- phaRNA Comprehensive RNA Technologies, Houston, Texas, USA
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nazish Sayed
- Department of Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Mark D Bonnen
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
43
|
Vanetti C, Vicentini LM, Cattaneo MG. Hormone-deprived serum impairs angiogenic properties in human endothelial cells regardless of estrogens. Endocr Res 2016; 41:325-333. [PMID: 27044317 DOI: 10.3109/07435800.2016.1155599] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS In vitro studies on hormone biological activities are commonly performed on cells cultured in nominally hormone-free media consisting of phenol-red-free media supplemented with charcoal-stripped (CS) serum. These media are largely used in almost all cell types, including endothelial cells (ECs). METHODS Cell number and metabolic activity were measured with standard methods. Angiogenesis was evaluated in a three-dimensional spheroid sprouting assay. RESULTS When we compared human umbilical vein ECs (HUVECs) cultured in standard conditions (199 medium supplemented with normal serum) with HUVECs grown in the hormone-free medium (phenol-red-free 199 medium supplemented with CS serum), we found that cells stop to grow in the absence of hormones. Notably, neither 17-β2 estradiol nor dihydrotestosterone reversed this inhibition. Moreover, the presence of the CS serum was sufficient to abrogate the ability of HUVECs to sprout in a three-dimensional spheroid assay, thus affecting a functional property of ECs. CONCLUSIONS Our results suggest that one or possibly more substances removed by stripping procedure from serum and different from sex hormones are crucial for the maintenance of in vitro ECs distinctive properties. Therefore, caution should be used when ECs are studied in media containing the CS serum.
Collapse
Affiliation(s)
- Claudia Vanetti
- a Department of Medical Biotechnology and Translational Medicine , Università degli Studi di Milano , Milano , Italy
| | - Lucia M Vicentini
- a Department of Medical Biotechnology and Translational Medicine , Università degli Studi di Milano , Milano , Italy
| | - Maria Grazia Cattaneo
- a Department of Medical Biotechnology and Translational Medicine , Università degli Studi di Milano , Milano , Italy
| |
Collapse
|
44
|
Eivers SB, Kinsella BT. Regulated expression of the prostacyclin receptor (IP) gene by androgens within the vasculature: Combined role for androgens and serum cholesterol. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1333-51. [PMID: 27365208 DOI: 10.1016/j.bbagrm.2016.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/13/2016] [Accepted: 06/24/2016] [Indexed: 01/11/2023]
Abstract
The prostanoid prostacyclin plays a key cardioprotective role within the vasculature. There is increasing evidence that androgens may also confer cardioprotection but through unknown mechanisms. This study investigated whether the androgen dihydrotestosterone (DHT) may regulate expression of the prostacyclin/I prostanoid receptor or, in short, the IP in platelet-progenitor megakaryoblastic and vascular endothelial cells. DHT significantly increased IP mRNA and protein expression, IP-induced cAMP generation and promoter (PrmIP)-directed gene expression in all cell types examined. The androgen-responsive region was localised to a cis-acting androgen response element (ARE), which lies in close proximity to a functional sterol response element (SRE) within the core promoter. In normal serum conditions, DHT increased IP expression through classic androgen receptor (AR) binding to the functional ARE within the PrmIP. However, under conditions of low-cholesterol, DHT led to further increases in IP expression through an indirect mechanism involving AR-dependent upregulation of SCAP expression and enhanced SREBP1 processing & binding to the SRE within the PrmIP. Chromatin immunoprecipitation assays confirmed DHT-induced AR binding to the ARE in vivo in cells cultured in normal serum while, in conditions of low cholesterol, DHT led to increased AR and SREBP1 binding to the functional ARE and SRE cis-acting elements, respectively, within the core PrmIP resulting in further increases in IP expression. Collectively, these data establish that the human IP gene is under the transcriptional regulation of DHT, where this regulation is further influenced by serum-cholesterol levels. This may explain, in part, some of the protective actions of androgens within the vasculature.
Collapse
Affiliation(s)
- Sarah B Eivers
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - B Therese Kinsella
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
45
|
Grossini E, Raina G, Farruggio S, Camillo L, Molinari C, Mary D, Walker GE, Bona G, Vacca G, Moia S, Prodam F, Surico D. Intracoronary Des-Acyl Ghrelin Acutely Increases Cardiac Perfusion Through a Nitric Oxide-Related Mechanism in Female Anesthetized Pigs. Endocrinology 2016; 157:2403-15. [PMID: 27100620 DOI: 10.1210/en.2015-1922] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Des-acyl ghrelin (DAG), the most abundant form of ghrelin in humans, has been found to reduce arterial blood pressure and prevent cardiac and endothelial cell apoptosis. Despite this, data regarding its direct effect on cardiac function and coronary blood flow, as well as the related involvement of autonomic nervous system and nitric oxide (NO), are scarce. We therefore examined these issues using both in vivo and in vitro studies. In 20 anesthetized pigs, intracoronary 100 pmol/mL DAG infusion with a constant heart rate and aortic blood pressure, increased coronary blood flow and NO release, whereas reducing coronary vascular resistances (P < .05). Dose responses to DAG were evaluated in five pigs. No effects on cardiac contractility/relaxation or myocardial oxygen consumption were observed. Moreover, whereas the blockade of muscarinic cholinoceptors (n = 5) or α- and β-adrenoceptors (n = 5 each) did not abolish the observed responses, NO synthase inhibition (n = 5) prevented the effects of DAG on coronary blood flow and NO release. In coronary artery endothelial cells, DAG dose dependently increased NO release through cAMP signaling and ERK1/2, Akt, and p38 MAPK involvement as well as the phosphorylation of endothelial NO synthase. In conclusion, in anesthetized pigs, DAG primarily increased cardiac perfusion through the involvement of NO release. Moreover, the phosphorylation of ERK1/2 and Akt appears to play roles in eliciting the observed NO production in coronary artery endothelial cells.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Giulia Raina
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Serena Farruggio
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Lara Camillo
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Claudio Molinari
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - David Mary
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Gillian Elisabeth Walker
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Gianni Bona
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Giovanni Vacca
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Stefania Moia
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Flavia Prodam
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| | - Daniela Surico
- Laboratory Physiology/Experimental Surgery (E.G., G.R., S.F., L.C., C.M., D.M., G.V., D.S.), Department of Translational Medicine, and Pediatric Unit (G.E.W., G.B., S.M., F.P.), Department of Health Sciences, University E Piedmont "A. Avogadro," Azienda Ospedaliera Universitaria Maggiore della Carità, I-12800 Novara, Italy
| |
Collapse
|
46
|
Hashimoto M, Miyai N, Hattori S, Iwahara A, Utsumi M, Arita M, Takeshita T. Age and gender differences in the influences of eNOS T-786C polymorphism on arteriosclerotic parameters in general population in Japan. Environ Health Prev Med 2016; 21:274-82. [PMID: 27038349 DOI: 10.1007/s12199-016-0527-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/19/2016] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The influence of T-786C polymorphism in the promoter region of endothelial nitric oxide synthase (eNOS) on arteriosclerotic parameters by age and gender were examined. METHODS Brachial-ankle pulse wave velocity (baPWV), heart-rate adjusted augmentation index (AIx@75), pulse pressure (PP) and albumin-creatinine ratio (ACR) were assessed as arteriosclerotic parameters in addition to non-high-density lipoprotein cholesterol (non-HDL-C) to HDL-C (non-HDL-C/HDL-C) ratio in 1499 participants. T-786C polymorphism (rs2070744) was screened using a TaqMan allelic discrimination assay. Analyses of covariance were carried. RESULTS Women with the non-C allele showed significantly lower AIx@75 in participants aged <65 years and baPWV in participants aged ≥65 years than those with C allele. In contrast, men with the non-C allele showed significantly higher PP in participants aged <65 years, and higher ACR and non-HDL-C/HDL-C ratio in participants aged ≥65 years. In men on cholesterol-lowering medication, the non-C allele carriers showed significantly higher non-HDL-C compared to those in the C allele carriers. CONCLUSIONS eNOS T-786C polymorphism is significantly associated with arteriosclerotic parameters accompanied with age and gender differences, possibly involving antioxidative and/or endothelial signaling other than inflammatory signaling.
Collapse
Affiliation(s)
- Marowa Hashimoto
- Department of Public Health, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama City, Wakayama, 641-8509, Japan
| | - Nobuyuki Miyai
- Wakayama Medical University School of Health and Nursing Science, 580 Mikazura, Wakayama City, Wakayama, 641-0011, Japan
| | - Sonomi Hattori
- Wakayama Medical University School of Health and Nursing Science, 580 Mikazura, Wakayama City, Wakayama, 641-0011, Japan
| | - Akihiko Iwahara
- Wakayama Medical University School of Health and Nursing Science, 580 Mikazura, Wakayama City, Wakayama, 641-0011, Japan
| | - Miyoko Utsumi
- Wakayama Medical University School of Health and Nursing Science, 580 Mikazura, Wakayama City, Wakayama, 641-0011, Japan
| | - Mikio Arita
- Wakayama Medical University School of Health and Nursing Science, 580 Mikazura, Wakayama City, Wakayama, 641-0011, Japan
| | - Tatsuya Takeshita
- Department of Public Health, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama City, Wakayama, 641-8509, Japan.
| |
Collapse
|
47
|
Cai JJ, Wen J, Jiang WH, Lin J, Hong Y, Zhu YS. Androgen actions on endothelium functions and cardiovascular diseases. J Geriatr Cardiol 2016; 13:183-196. [PMID: 27168746 PMCID: PMC4854959 DOI: 10.11909/j.issn.1671-5411.2016.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 01/12/2016] [Accepted: 01/19/2016] [Indexed: 12/02/2022] Open
Abstract
The roles of androgens on cardiovascular physiology and pathophysiology are controversial as both beneficial and detrimental effects have been reported. Although the reasons for this discrepancy are unclear, multiple factors such as genetic and epigenetic variation, sex-specificity, hormone interactions, drug preparation and route of administration may contribute. Recently, growing evidence suggests that androgens exhibit beneficial effects on cardiovascular function though the mechanism remains to be elucidated. Endothelial cells (ECs) which line the interior surface of blood vessels are distributed throughout the circulatory system, and play a crucial role in cardiovascular function. Endothelial progenitor cells (EPCs) are considered an indispensable element for the reconstitution and maintenance of an intact endothelial layer. Endothelial dysfunction is regarded as an initiating step in development of atherosclerosis and cardiovascular diseases. The modulation of endothelial functions by androgens through either genomic or nongenomic signal pathways is one possible mechanism by which androgens act on the cardiovascular system. Obtaining insight into the mechanisms by which androgens affect EC and EPC functions will allow us to determine whether androgens possess beneficial effects on the cardiovascular system. This in turn may be critical in the prevention and therapy of cardiovascular diseases. This article seeks to review recent progress in androgen regulation of endothelial function, the sex-specificity of androgen actions, and its clinical applications in the cardiovascular system.
Collapse
Affiliation(s)
- Jing-Jing Cai
- Department of Cardiology of the Third Xiangya Hospital, Central South University, Changsha, China; The Center of Clinical Pharmacology of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Juan Wen
- Department of Cardiology of the Third Xiangya Hospital, Central South University, Changsha, China; The Center of Clinical Pharmacology of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Hong Jiang
- Department of Cardiology of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Jian Lin
- Department of Medicine/Endocrinology, Weill Cornell Medical College, NY, USA
| | - Yuan Hong
- Department of Cardiology of the Third Xiangya Hospital, Central South University, Changsha, China; The Center of Clinical Pharmacology of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuan-Shan Zhu
- Department of Medicine/Endocrinology, Weill Cornell Medical College, NY, USA; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Yoshida S, Ikeda Y, Aihara KI. Roles of the Androgen – Androgen Receptor System in Vascular Angiogenesis. J Atheroscler Thromb 2016; 23:257-65. [DOI: 10.5551/jat.31047] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Sumiko Yoshida
- Department of Hematology, Endocrinology and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School
| | - Ken-ichi Aihara
- Department of Hematology, Endocrinology and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School
| |
Collapse
|
49
|
Musicki B, Bella AJ, Bivalacqua TJ, Davies KP, DiSanto ME, Gonzalez-Cadavid NF, Hannan JL, Kim NN, Podlasek CA, Wingard CJ, Burnett AL. Basic Science Evidence for the Link Between Erectile Dysfunction and Cardiometabolic Dysfunction. J Sex Med 2015; 12:2233-55. [PMID: 26646025 DOI: 10.1111/jsm.13069] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Although clinical evidence supports an association between cardiovascular/metabolic diseases (CVMD) and erectile dysfunction (ED), scientific evidence for this link is incompletely elucidated. AIM This study aims to provide scientific evidence for the link between CVMD and ED. METHODS In this White Paper, the Basic Science Committee of the Sexual Medicine Society of North America assessed the current literature on basic scientific support for a mechanistic link between ED and CVMD, and deficiencies in this regard with a critical assessment of current preclinical models of disease. RESULTS A link exists between ED and CVMD on several grounds: the endothelium (endothelium-derived nitric oxide and oxidative stress imbalance); smooth muscle (SM) (SM abundance and altered molecular regulation of SM contractility); autonomic innervation (autonomic neuropathy and decreased neuronal-derived nitric oxide); hormones (impaired testosterone release and actions); and metabolics (hyperlipidemia, advanced glycation end product formation). CONCLUSION Basic science evidence supports the link between ED and CVMD. The Committee also highlighted gaps in knowledge and provided recommendations for guiding further scientific study defining this risk relationship. This endeavor serves to develop novel strategic directions for therapeutic interventions.
Collapse
Affiliation(s)
- Biljana Musicki
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Anthony J Bella
- Division of Urology, Department of Surgery and Department of Neuroscience, Ottawa Hospital Research Institute at the University of Ottawa, Ottawa, ON, Canada
| | - Trinity J Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kelvin P Davies
- Department of Urology, Albert Einstein College of Medicine, New York, NY, USA
| | - Michael E DiSanto
- Department of Surgery/Division of Urology, Cooper University Hospital, Camden, NJ, USA
| | - Nestor F Gonzalez-Cadavid
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA.,Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Johanna L Hannan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Noel N Kim
- Institute for Sexual Medicine, San Diego, CA, USA
| | - Carol A Podlasek
- Departments of Urology, Physiology, and Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Christopher J Wingard
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Arthur L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Tilakaratne A, Soory M. Antioxidant response of osteoblasts to doxycycline in an inflammatory model induced by C-reactive protein and interleukin-6. Infect Disord Drug Targets 2015; 14:14-22. [PMID: 25159306 PMCID: PMC4443794 DOI: 10.2174/1871526514666140827101231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 12/15/2022]
Abstract
Objectives: Investigation of osteoblastic responses to oxidative stress, induced by C-reactive protein (CRP) and IL-6 and ameliorating effects of doxycycline (Dox); using assays for 5-alpha dihydrotestosterone (DHT) as an antioxidant marker of healing. IL-6 and CRP are risk markers of periodontitis and prevalent comorbidities in periodontitis subjects. Methods: Confluent monolayer cultures of osteoblasts were incubated with radiolabelled testosterone (14C-T) as substrate, in the presence or absence (Control) of pre-determined optimal concentrations of CRP, IL-6, Dox; alone and in combination (n=8) for 24h in MEM. The eluent was solvent-extracted for steroid metabolites. They were separated using TLC in a benzene/ acetone solvent system 4:1 v/v; and quantified using radioisotope scanning. The identity of formed metabolites was confirmed using the mobility of cold standards added to the samples and disclosed in iodine. Further confirmation of the authenticity of DHT was carried out by combined gas chromatrography-mass spectrometry, after derivatization to pentafluorobenzyloxime trimethyl silyl ether. Results: The yields of DHT from 14C-testosterone showed 2-fold and 1.8-fold- inhibition in response to IL-6 and CRP respectively and 28% stimulation in response to Dox, via the 5-alpha reductase pathway. The combination of IL-6 + CRP showed a 2-fold reduction in the yields of DHT, elevated to control values when combined with Dox (n=8; p<0.001). Yields of 4-androstenedione showed an inverse relationship to those of DHT, in response to the agents tested, in keeping with the 17-beta hydroxysteroid dehydrogenase pathway. Conclusions: Inhibition of DHT synthesis in osteoblasts by IL-6 and CRP was overcome by doxycycline. Oxidative actions of IL-6 and CRP; and antioxidant actions of Dox are reinforced by the metabolic yields of DHT in response to agents tested. Using a novel metabolically active model allows closer extrapolation to in vivo conditions; in the context of adjunctive therapeutic applications for periodontitis and prevalent comorbidities.
Collapse
Affiliation(s)
| | - Mena Soory
- King's College London Dental Institute, Denmark Hill, London SE5 9RW, UK.
| |
Collapse
|