1
|
Anger JT, Case LK, Baranowski AP, Berger A, Craft RM, Damitz LA, Gabriel R, Harrison T, Kaptein K, Lee S, Murphy AZ, Said E, Smith SA, Thomas DA, Valdés Hernández MDC, Trasvina V, Wesselmann U, Yaksh TL. Pain mechanisms in the transgender individual: a review. FRONTIERS IN PAIN RESEARCH 2024; 5:1241015. [PMID: 38601924 PMCID: PMC11004280 DOI: 10.3389/fpain.2024.1241015] [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: 06/15/2023] [Accepted: 01/25/2024] [Indexed: 04/12/2024] Open
Abstract
Specific Aim Provide an overview of the literature addressing major areas pertinent to pain in transgender persons and to identify areas of primary relevance for future research. Methods A team of scholars that have previously published on different areas of related research met periodically though zoom conferencing between April 2021 and February 2023 to discuss relevant literature with the goal of providing an overview on the incidence, phenotype, and mechanisms of pain in transgender patients. Review sections were written after gathering information from systematic literature searches of published or publicly available electronic literature to be compiled for publication as part of a topical series on gender and pain in the Frontiers in Pain Research. Results While transgender individuals represent a significant and increasingly visible component of the population, many researchers and clinicians are not well informed about the diversity in gender identity, physiology, hormonal status, and gender-affirming medical procedures utilized by transgender and other gender diverse patients. Transgender and cisgender people present with many of the same medical concerns, but research and treatment of these medical needs must reflect an appreciation of how differences in sex, gender, gender-affirming medical procedures, and minoritized status impact pain. Conclusions While significant advances have occurred in our appreciation of pain, the review indicates the need to support more targeted research on treatment and prevention of pain in transgender individuals. This is particularly relevant both for gender-affirming medical interventions and related medical care. Of particular importance is the need for large long-term follow-up studies to ascertain best practices for such procedures. A multi-disciplinary approach with personalized interventions is of particular importance to move forward.
Collapse
Affiliation(s)
- Jennifer T. Anger
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Laura K. Case
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| | - Andrew P. Baranowski
- Pelvic Pain Medicine and Neuromodulation, University College Hospital Foundation Trust, University College London, London, United Kingdom
| | - Ardin Berger
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Rebecca M. Craft
- Department of Psychology, Washington State University, Pullman, WA, United States
| | - Lyn Ann Damitz
- Division of Plastic and Reconstructive Surgery, University of North Carolina, Chapel Hill, NC, United States
| | - Rodney Gabriel
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Tracy Harrison
- Department of OB/GYN & Reproductive Sciences, University of California San Diego, San Diego, CA, United States
| | - Kirsten Kaptein
- Division of Plastic Surgery, University of California San Diego, San Diego, CA, United States
| | - Sanghee Lee
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Anne Z. Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Engy Said
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Stacey Abigail Smith
- Division of Infection Disease, The Hope Clinic of Emory University, Atlanta, GA, United States
| | - David A. Thomas
- Office of Research on Women's Health, National Institutes of Health, Bethesda, MD, United States
| | - Maria del C. Valdés Hernández
- Department of Neuroimaging Sciences, Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor Trasvina
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Ursula Wesselmann
- Departments of Anesthesiology and Perioperative Medicine/Division of Pain Medicine, Neurology and Psychology, and Consortium for Neuroengineering and Brain-Computer Interfaces, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
2
|
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
|
3
|
Mascone SE, Kim KI, Evans WS, Prior SJ, Cook MD, Ranadive SM. Race and sex differences in ROS production and SOD activity in HUVECs. PLoS One 2023; 18:e0292112. [PMID: 37792791 PMCID: PMC10550108 DOI: 10.1371/journal.pone.0292112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Black individuals and men are predisposed to an earlier onset and higher prevalence of hypertension, compared with White individuals and women, respectively. Therefore, the influence of race and sex on reactive oxygen species (ROS) production and superoxide dismutase (SOD) activity following induced inflammation was evaluated in female and male human umbilical vein endothelial cells (HUVECs) from Black and White individuals. It was hypothesized that HUVECs from Black individuals and male HUVECs would exhibit greater ROS production and impaired SOD activity. Inflammation was induced in HUVEC cell lines (n = 4/group) using tumor necrosis factor-alpha (TNF-α, 50ng/ml). There were no between group differences in ROS production or SOD activity in HUVECs from Black and White individuals, and HUVECs from Black individuals exhibited similar SOD activity at 24hr compared with 4hr of TNF-α treatment (p>0.05). However, HUVECs from White individuals exhibited significantly greater SOD Activity (p<0.05) at 24hr as compared to 4hr in the control condition but not with TNF-α treatment (p>0.05). Female HUVECs exhibited significantly lower ROS production than male HUVECs in the control condition and following TNF-α induced inflammation (p<0.05). Only female HUVECs exhibited significant increases in SOD activity with increased exposure time to TNF-α induced inflammation (p<0.05). HUVECs from White individuals alone exhibit blunted SOD activity when comparing control and TNF-α conditions. Further, compared to female HUVECs, male HUVECs exhibit a pro-inflammatory state.
Collapse
Affiliation(s)
- Sara E. Mascone
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, United States of America
| | - Katherine I. Kim
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, United States of America
| | - William S. Evans
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, United States of America
| | - Steven J. Prior
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, United States of America
| | - Marc D. Cook
- Department of Kinesiology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, United States of America
| | - Sushant M. Ranadive
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, United States of America
| |
Collapse
|
4
|
Martier AT, Maurice YV, Conrad KM, Mauvais-Jarvis F, Mondrinos MJ. Sex-specific actions of estradiol and testosterone on human fibroblast and endothelial cell proliferation, bioenergetics, and vasculogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550236. [PMID: 37546849 PMCID: PMC10402012 DOI: 10.1101/2023.07.23.550236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Progress toward the development of sex-specific tissue engineered systems has been hampered by the lack of research efforts to define the effects of sex-specific hormone concentrations on relevant human cell types. Here, we investigated the effects of defined concentrations of estradiol (E2) and dihydrotestosterone (DHT) on primary human dermal and lung fibroblasts (HDF and HLF), and human umbilical vein endothelial cells (HUVEC) from female (XX) and male (XY) donors in both 2D expansion cultures and 3D stromal vascular tissues. Sex-matched E2 and DHT stimulation in 2D expansion cultures significantly increased the proliferation index, mitochondrial membrane potential, and the expression of genes associated with bioenergetics (Na+/K+ ATPase, somatic cytochrome C) and beneficial stress responses (chaperonin) in all cell types tested. Notably, cross sex hormone stimulation, i.e., DHT treatment of XX cells in the absence of E2 and E2 stimulation of XY cells in the absence of DHT, decreased bioenergetic capacity and inhibited cell proliferation. We used a microengineered 3D vasculogenesis assay to assess hormone effects on tissue scale morphogenesis. E2 increased metrics of vascular network complexity compared to vehicle in XX tissues. Conversely, and in line with results from 2D expansion cultures, E2 potently inhibited vasculogenesis compared to vehicle in XY tissues. DHT did not significantly alter vasculogenesis in XX or XY tissues but increased the number of non-participating endothelial cells in both sexes. This study establishes a scientific rationale and adaptable methods for using sex hormone stimulation to develop sex-specific culture systems.
Collapse
Affiliation(s)
- Ashley T. Martier
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Yasmin V. Maurice
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - K. Michael Conrad
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Franck Mauvais-Jarvis
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Section of Endocrinology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Southeast Louisiana VA Medical Center, New Orleans, LA, USA
| | - Mark J. Mondrinos
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
5
|
Satam K, Ohashi Y, Thaxton C, Gonzalez L, Setia O, Bai H, Aoyagi Y, Xie Y, Zhang W, Yatsula B, Martin KA, Cai Y, Dardik A. Sex hormones impact early maturation and immune response in the arteriovenous fistula mouse model. Am J Physiol Heart Circ Physiol 2023; 325:H77-H88. [PMID: 37145957 PMCID: PMC10243550 DOI: 10.1152/ajpheart.00049.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/07/2023]
Abstract
Arteriovenous fistulae (AVF) fail to mature more frequently in female patients compared with male patients, leading to inferior outcomes and decreased utilization. Since our mouse AVF model recapitulates sex differences in human AVF maturation, we hypothesized that sex hormones mediate these differences during AVF maturation. C57BL/6 mice (9-11 wk) were treated with aortocaval AVF surgery and/or gonadectomy. AVF hemodynamics were measured via ultrasound (days 0-21). Blood was collected for FACS and tissue for immunofluorescence and ELISA (days 3 and 7); wall thickness was assessed by histology (day 21). Inferior vena cava shear stress was higher in male mice (P = 0.0028) after gonadectomy, and they had increased wall thickness (22.0 ± 1.8 vs. 12.7 ± 1.2 µm; P < 0.0001). Conversely, female mice had decreased wall thickness (6.8 ± 0.6 vs. 15.3 ± 0.9 µm; P = 0.0002). Intact female mice had higher proportions of circulating CD3+ T cells on day 3 (P = 0.0043), CD4+ (P = 0.0003) and CD8+ T cells (P = 0.005) on day 7, and CD11b+ monocytes on day 3 (P = 0.0046). After gonadectomy, these differences disappeared. In intact female mice, CD3+ T cells (P = 0.025), CD4+ T cells (P = 0.0178), CD8+ T cells (P = 0.0571), and CD68+ macrophages (P = 0.0078) increased in the fistula wall on days 3 and 7. This disappeared after gonadectomy. Furthermore, female mice had higher IL-10 (P = 0.0217) and TNF-α (P = 0.0417) levels in their AVF walls than male mice. Sex hormones mediate AVF maturation, suggesting that hormone receptor signaling may be a target to improve AVF maturation.NEW & NOTEWORTHY After arteriovenous fistula creation, females have lower rates of maturation and higher rates of failure than males. In a mouse model of venous adaptation that recapitulates human fistula maturation, sex hormones may be mechanisms of the sexual dimorphism: testosterone is associated with reduced shear stress, whereas estrogen is associated with increased immune cell recruitment. Modulating sex hormones or downstream effectors suggests sex-specific therapies and could address disparities in sex differences in clinical outcomes.
Collapse
Affiliation(s)
- Keyuree Satam
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Carly Thaxton
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States
| | - Luis Gonzalez
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ocean Setia
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yangzhouyun Xie
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Kathleen A Martin
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yujun Cai
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
6
|
Abstract
Although sex differences have been noted in cellular function and behavior, therapy efficacy, and disease incidence and outcomes, the adoption of sex as a biological variable in tissue engineering and regenerative medicine remains limited. Furthering the development of personalized, precision medicine requires considering biological sex at the bench and in the clinic. This review provides the basis for considering biological sex when designing tissue-engineered constructs and regenerative therapies by contextualizing sex as a biological variable within the tissue engineering triad of cells, matrices, and signals. To achieve equity in biological sex within medicine requires a cultural shift in science and engineering research, with active engagement by researchers, clinicians, companies, policymakers, and funding agencies.
Collapse
Affiliation(s)
- Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Christopher Ludtka
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Bryan D James
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA;
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA
| |
Collapse
|
7
|
Willemars MMA, Nabben M, Verdonschot JAJ, Hoes MF. Evaluation of the Interaction of Sex Hormones and Cardiovascular Function and Health. Curr Heart Fail Rep 2022; 19:200-212. [PMID: 35624387 PMCID: PMC9329157 DOI: 10.1007/s11897-022-00555-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 12/02/2022]
Abstract
Purpose of Review Sex hormones drive development and function of reproductive organs or the development of secondary sex characteristics but their effects on the cardiovascular system are poorly understood. In this review, we identify the gaps in our understanding of the interaction between sex hormones and the cardiovascular system. Recent Findings Studies are progressively elucidating molecular functions of sex hormones in specific cell types in parallel with the initiation of crucial large randomized controlled trials aimed at improving therapies for cardiovascular diseases (CVDs) associated with aberrant levels of sex hormones. Summary In contrast with historical assumptions, we now understand that men and women show different symptoms and progression of CVDs. Abnormal levels of sex hormones pose an independent risk for CVD, which is apparent in conditions like Klinefelter syndrome, androgen insensitivity syndrome, and menopause. Moreover, sex hormone–based therapies remain understudied and may not be beneficial for cardiovascular health.
Collapse
Affiliation(s)
- Myrthe M A Willemars
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands.,Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Job A J Verdonschot
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands.,Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Martijn F Hoes
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands. .,Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands. .,Department of Cardiology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
8
|
Lin HL, Lee CY, Huang JY, Tseng PC, Yang SF. Androgen Deprivation Therapy for Prostate Cancer Did Not Increase the Risk of Retinal Vascular Occlusion: A Population-Based Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042268. [PMID: 35206456 PMCID: PMC8871724 DOI: 10.3390/ijerph19042268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 11/16/2022]
Abstract
This study aimed to evaluate the effect of androgen deprivation therapy (ADT) on retinal vascular occlusion (RVO) development in patients with prostate cancer, using data from Taiwan’s National Health Insurance Research Database. A total of 1791, 1791, and 3582 patients were enrolled in the prostate cancer with ADT group, prostate cancer without ADT group, and the control group, respectively. The primary outcome was RVO occurrence, according to diagnostic codes. Cox proportional hazard regression was used to determine the adjusted hazard ratio (aHR) and 95% confidence interval (CI) of ADT and other covariates for RVO incidence. After a follow-up interval of up to 18 years, the patients with prostate cancer who received ADT showed significantly lower RVO incidence than the control group (aHR: 0.191, 95% CI: 0.059–0.621, p = 0.0059), after adjusting for multiple confounders. Hypertension was related to higher RVO incidence (aHR: 2.130, 95% CI: 1.127–4.027, p = 0.0199). Our overall results showed that using ADT for prostate cancer did not lead to a greater risk of RVO development. In fact, the patients with prostate cancer who received ADT had lower RVO incidence than those who did not receive ADT.
Collapse
Affiliation(s)
- Hsin-Le Lin
- Department of Ophthalmology, Taipei City Hospital, Renai Branch, Taipei 106243, Taiwan;
| | - Chia-Yi Lee
- Department of Ophthalmology, Nobel Eye Institute, Taipei 106074, Taiwan;
| | - Jing-Yang Huang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan;
| | - Po-Chen Tseng
- Department of Ophthalmology, Taipei City Hospital, Renai Branch, Taipei 106243, Taiwan;
- Department of Special Education, University of Taipei, Taipei 100234, Taiwan
- Department of Optometry, University of Kang-Ning, Taipei 114311, Taiwan
- Correspondence: (P.-C.T.); (S.-F.Y.); Tel.: +886-2-2709-3600 (ext. 3395) (P.-C.T.); +886-4-24739595 (ext. 34253) (S.-F.Y.)
| | - Shun-Fa Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan;
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan
- Correspondence: (P.-C.T.); (S.-F.Y.); Tel.: +886-2-2709-3600 (ext. 3395) (P.-C.T.); +886-4-24739595 (ext. 34253) (S.-F.Y.)
| |
Collapse
|
9
|
Leach DA, Fernandes RC, Bevan CL. Cellular specificity of androgen receptor, coregulators, and pioneer factors in prostate cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2022; 2:R112-R131. [PMID: 37435460 PMCID: PMC10259329 DOI: 10.1530/eo-22-0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 07/13/2023]
Abstract
Androgen signalling, through the transcription factor androgen receptor (AR), is vital to all stages of prostate development and most prostate cancer progression. AR signalling controls differentiation, morphogenesis, and function of the prostate. It also drives proliferation and survival in prostate cancer cells as the tumour progresses; given this importance, it is the main therapeutic target for disseminated disease. AR is also essential in the surrounding stroma, for the embryonic development of the prostate and controlling epithelial glandular development. Stromal AR is also important in cancer initiation, regulating paracrine factors that excite cancer cell proliferation, but lower stromal AR expression correlates with shorter time to progression/worse outcomes. The profile of AR target genes is different between benign and cancerous epithelial cells, between castrate-resistant prostate cancer cells and treatment-naïve cancer cells, between metastatic and primary cancer cells, and between epithelial cells and fibroblasts. This is also true of AR DNA-binding profiles. Potentially regulating the cellular specificity of AR binding and action are pioneer factors and coregulators, which control and influence the ability of AR to bind to chromatin and regulate gene expression. The expression of these factors differs between benign and cancerous cells, as well as throughout disease progression. The expression profile is also different between fibroblast and mesenchymal cell types. The functional importance of coregulators and pioneer factors in androgen signalling makes them attractive therapeutic targets, but given the contextual expression of these factors, it is essential to understand their roles in different cancerous and cell-lineage states.
Collapse
Affiliation(s)
- Damien A Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Rayzel C Fernandes
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
10
|
Converse A, Thomas P. Androgens promote vascular endothelial cell proliferation through activation of a ZIP9-dependent inhibitory G protein/PI3K-Akt/Erk/cyclin D1 pathway. Mol Cell Endocrinol 2021; 538:111461. [PMID: 34555425 DOI: 10.1016/j.mce.2021.111461] [Citation(s) in RCA: 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: 05/16/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022]
Abstract
While androgens have been reported to mediate cardiovascular endothelial cell proliferation, the potential involvement of membrane androgen receptors (mAR) has not been examined. Here we show ZIP9, a recently characterized mAR, mediates androgen-induced early proliferative events in human umbilical vein endothelial cells (HUVECs). Androgen treatment significantly increased cyclin D1 nuclear localization and proliferation, which were blocked by transfection with siRNA targeting ZIP9 but not the nuclear AR. Testosterone rapidly activated inhibitory G protein signaling, Erk, and Akt, and inhibition of these signaling members abrogated the ZIP9-mediated cyclin D1 and proliferative responses. Erk and Akt modulated cyclin D1 nuclear localization by upregulation of cyclin D1 mRNA and inhibition of GSK-3β activity, respectively. This is the first study to demonstrate a role for ZIP9 in HUVEC proliferation and indicates ZIP9 is a physiologically-relevant androgen receptor in the cardiovascular system that merits further study as a potential therapeutic target for treating cardiovascular disease.
Collapse
Affiliation(s)
- Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, Port Aransas, TX, USA.
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, Port Aransas, TX, USA
| |
Collapse
|
11
|
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: 1.3] [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
|
12
|
James BD, Allen JB. Sex-Specific Response to Combinations of Shear Stress and Substrate Stiffness by Endothelial Cells In Vitro. Adv Healthc Mater 2021; 10:e2100735. [PMID: 34142471 PMCID: PMC8458248 DOI: 10.1002/adhm.202100735] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 12/25/2022]
Abstract
By using a full factorial design of experiment, the combinatorial effects of biological sex, shear stress, and substrate stiffness on human umbilical vein endothelial cell (HUVEC) spreading and Yes-associated protein 1 (YAP1) activity are able to be efficiently evaluated. Within the range of shear stress (0.5-1.5 Pa) and substrate stiffness (10-100 kPa), male HUVECs are smaller than female HUVECs. Only with sufficient mechanical stimulation do they spread to a similar size. More importantly, YAP1 nuclear localization in female HUVECs is invariant to mechanical stimulation within the range of tested conditions whereas for male HUVECs it increases nonlinearly with increasing shear stress and substrate stiffness. The sex-specific response of HUVECs to combinations of shear stress and substrate stiffness reinforces the need to include sex as a biological variable and multiple mechanical stimuli in experiments, informs the design of precision biomaterials, and offers insight for understanding cardiovascular disease sexual dimorphisms. Moreover, here it is illustrated that different complex mechanical microenvironments can lead to sex-specific phenotypes and sex invariant phenotypes in cultured endothelial cells.
Collapse
Affiliation(s)
- Bryan D James
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| |
Collapse
|
13
|
Nasser SA, Afify EA, Kobeissy F, Hamam B, Eid AH, El-Mas MM. Inflammatory Basis of Atherosclerosis: Modulation by Sex Hormones. Curr Pharm Des 2021; 27:2099-2111. [PMID: 33480335 DOI: 10.2174/1381612827666210122142811] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/17/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis-related cardiovascular diseases (CVDs) are the leading cause of death globally. Several lines of evidence are supportive of the contributory role of vascular inflammation in atherosclerosis. Diverse immune cell types, including monocytes/macrophages, T-cells and neutrophils, as well as specialized proresolving lipid mediators, have been successfully characterized as key players in vascular inflammation. The increased prevalence of atherosclerotic CVD in men in comparison to age-matched premenopausal women and the abolition of sex differences in prevalence during menopause strongly suggest a pivotal role of sex hormones in the development of CVD. Indeed, many animal and human studies conclusively implicate sex hormones as a crucial component in driving the immune response. This is further corroborated by the effective identification of sex hormone receptors in vascular endothelial cells, vascular smooth muscle cells and immune cells. Collectively, these findings suggest a cellular communication between sex hormones and vascular or immune cells underlying the vascular inflammation in atherosclerosis. The aim of this review is to provide an overview of vascular inflammation as a causal cue underlying atherosclerotic CVDs within the context of the modulatory effects of sex hormones. Moreover, the cellular and molecular signaling pathways underlying the sex hormones- immune system interactions as potential culprits for vascular inflammation are highlighted with detailed and critical discussion. Finally, the review concludes by speculations on the potential sex-related efficacy of currently available immunotherapies in mitigating vascular inflammation. Conceivably, a deeper understanding of the immunoregulatory influence of sex hormones on vascular inflammation-mediated atherosclerosis permits sex-based management of atherosclerosis-related CVDs.
Collapse
Affiliation(s)
- Suzanne A Nasser
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, P.O. Box 11-5020, Beirut, Lebanon
| | - Elham A Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Bassam Hamam
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, P.O. Box 146404, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
14
|
Chan SM, Weininger G, Langford J, Jane-Wit D, Dardik A. Sex Differences in Inflammation During Venous Remodeling of Arteriovenous Fistulae. Front Cardiovasc Med 2021; 8:715114. [PMID: 34368264 PMCID: PMC8335484 DOI: 10.3389/fcvm.2021.715114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular disorders frequently have differing clinical presentations among women and men. Sex differences exist in vascular access for hemodialysis; women have reduced rates of arteriovenous fistula (AVF) maturation as well as fistula utilization compared with men. Inflammation is increasingly implicated in both clinical studies and animal models as a potent mechanism driving AVF maturation, especially in vessel dilation and wall thickening, that allows venous remodeling to the fistula environment to support hemodialysis. Sex differences have long been recognized in arterial remodeling and diseases, with men having increased cardiovascular events compared with pre-menopausal women. Many of these arterial diseases are driven by inflammation that is similar to the inflammation during AVF maturation. Improved understanding of sex differences in inflammation during vascular remodeling may suggest sex-specific vascular therapies to improve AVF success.
Collapse
Affiliation(s)
- Shin Mei Chan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - Gabe Weininger
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Daniel Jane-Wit
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Veterans Affairs (VA) Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
15
|
Sharmeen S, Nomani H, Taub E, Carlson H, Yao Q. Polycystic ovary syndrome: epidemiologic assessment of prevalence of systemic rheumatic and autoimmune diseases. Clin Rheumatol 2021; 40:4837-4843. [PMID: 34216315 DOI: 10.1007/s10067-021-05850-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/20/2021] [Accepted: 06/27/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) causes anovulation and hyperandrogenism. Hormonal imbalance is known to contribute to systemic autoimmune diseases. OBJECTIVE To examine the frequency of certain rheumatic diseases in PCOS. METHODS This retrospective study utilized and analyzed electronic medical records from January 2004 through February 2020. A diagnosis of PCOS and specified rheumatic diseases was searched using ICD-9 and ICD-10 codes. A total of 754 adult patients with PCOS and 1,508 age- and body mass index-matched patients without PCOS were included. Frequencies of the rheumatic diseases were compared between PCOS and non-PCOS subjects or literature data. RESULTS The prevalence of rheumatoid arthritis (RA) was found to be 2.25% (17/737) in the PCOS patients, numerically higher than 1.26% (19/1489) in the non-PCOS subjects. The difference was significant with a confidence level of 90% (1.04-3.15) but not at 95% with an odds ratio of 1.808 (95% CI = 0.934-3.4, p = 0.0747). When compared with the literature data from the US female population, the prevalence of RA in PCOS patients was significantly higher (2.25% vs. 1.40%, p < 0.0001). Among the autoimmune diseases examined, both systemic sclerosis (0.40% vs. 0.0%, p = 0.0369) and undifferentiated connective tissue disease (0.53% vs. 0.0%, p = 0.0123) were significantly more frequent in the PCOS patients than the non-PCOS. Additionally, PCOS patients had a significantly higher frequency of osteoarthritis than non-PCOS patients (5.44% vs. 2.92%, p = 0.0030) with an odds ratio of 1.913 (95% CI = 1.239-2.955). CONCLUSION We have shown unprecedentedly that certain rheumatic diseases are more prevalent in PCOS. This study provides important insight into autoimmunity in association with PCOS. Key Points • Polycystic ovary syndrome is postulated to cause systemic autoimmune disease due to its hormonal imbalance. • We conducted the first epidemiologic assessment of the prevalence of systemic autoimmune diseases. • Certain autoimmune and rheumatic diseases are more prevalent in polycystic ovary syndrome.
Collapse
Affiliation(s)
- Saika Sharmeen
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Hafsa Nomani
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Erin Taub
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Harold Carlson
- Division of Endocrinology, Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Qingping Yao
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA.
| |
Collapse
|
16
|
Xia J, Gu L, Guo Y, Feng H, Chen S, Jurat J, Fu W, Zhang D. Gut Microbiota Mediates the Preventive Effects of Dietary Capsaicin Against Depression-Like Behavior Induced by Lipopolysaccharide in Mice. Front Cell Infect Microbiol 2021; 11:627608. [PMID: 33987106 PMCID: PMC8110911 DOI: 10.3389/fcimb.2021.627608] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 12/26/2022] Open
Abstract
Capsaicin (CAP) is an active ingredient in chili pepper that is frequently consumed. It exerts various pharmacological activities, and also has potential effects on mental illness. However, its mechanism of antidepressant effects is still unclear. Based on the emerging perspective of the gut-brain axis, we investigated the effects of dietary CAP on gut microbes in mice with depression-like behaviors induced by lipopolysaccharide (LPS). C57BL/6J male mice (four weeks old) were given specific feed (standard laboratory chow or laboratory chow plus 0.005% CAP) for 4 months. During the last five days, LPS (0.052/0.104/0.208/0.415/0.83 mg/kg, 5-day) was injected intraperitoneally to induce depression. Behavioral indicators and serum parameters were measured, and gut microbiota were identified by sequencing analysis of the 16S gene. This study showed that dietary CAP improved depressive-like behavior (sucrose preference test, forced swimming test, tail suspension test) and levels of 5-HT and TNF-α in serum of LPS-induced mice with depression-like behaviors. In addition, CAP could recover abnormal changes in depression-related microbiota. Especially at the genus level, CAP enhanced the variations in relative abundance of certain pivotal microorganisms like Ruminococcus, Prevotella, Allobaculum, Sutterella, and Oscillospira. Correlation analysis revealed changes in microbiota composition that was closely related to depressive behavior, 5-HT and TNF-α levels. These results suggested that dietary CAP can regulate the structure and number of gut microbiota and play a major role in the prevention of depression.
Collapse
Affiliation(s)
- Jing Xia
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Li Gu
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Yitong Guo
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Hongyan Feng
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Shuhan Chen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Jessore Jurat
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Wenjing Fu
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Dongfang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
17
|
Human Umbilical Cord: Information Mine in Sex-Specific Medicine. Life (Basel) 2021; 11:life11010052. [PMID: 33451112 PMCID: PMC7828611 DOI: 10.3390/life11010052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Biological differences between sexes should be considered in all stages of research, as sexual dimorphism starts in utero leading to sex-specific fetal programming. In numerous biomedical fields, there is still a lack of stratification by sex despite primary cultured cells retaining memory of the sex and of the donor. The sex of donors in biological research must be known because variations in cells and cellular components can be used as endpoints, biomarkers and/or targets of pharmacological studies. This selective review focuses on the current findings regarding sex differences observed in the umbilical cord, a widely used source of research samples, both in the blood and in the circulating cells, as well as in the different cellular models obtainable from it. Moreover, an overview on sex differences in fetal programming is reported. As it emerges that the sex variable is still often forgotten in experimental models, we suggest that it should be mandatory to adopt sex-oriented research, because only awareness of these issues can lead to innovative research.
Collapse
|
18
|
Boscaro C, Trenti A, Baggio C, Scapin C, Trevisi L, Cignarella A, Bolego C. Sex Differences in the Pro-Angiogenic Response of Human Endothelial Cells: Focus on PFKFB3 and FAK Activation. Front Pharmacol 2020; 11:587221. [PMID: 33390959 PMCID: PMC7773665 DOI: 10.3389/fphar.2020.587221] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/06/2020] [Indexed: 01/14/2023] Open
Abstract
Female hormones and sex-specific factors are established determinants of endothelial function, yet their relative contribution to human endothelium phenotypes has not been defined. Using human umbilical vein endothelial cells (HUVECs) genotyped by donor's sex, we investigated the influence of sex and estrogenic agents on the main steps of the angiogenic process and on key proteins governing HUVEC metabolism and migratory properties. HUVECs from female donors (fHUVECs) showed increased viability (p < 0.01) and growth rate (p < 0.01) compared with those from males (mHUVECs). Despite higher levels of G-protein coupled estrogen receptor (GPER) in fHUVECs (p < 0.001), treatment with 17β-estradiol (E2) and the selective GPER agonist G1 (both 1-100 nM) did not affect HUVEC viability. Migration and tubularization in vitro under physiological conditions were higher in fHUVECs than in mHUVECs (p < 0.05). E2 treatment (1-100 nM) upregulated the glycolytic activator PFKFB3 with higher potency in fHUVECs than in mHUVECs, despite comparable baseline levels. Moreover, Y576/577 phosphorylation of focal adhesion kinase (FAK) was markedly enhanced in fHUVECs (p < 0.001), despite comparable Src activation levels. While the PI3K inhibitor LY294002 (25 µM) inhibited HUVEC migration (p < 0.05), Akt phosphorylation levels in fHUVECs and mHUVECs were comparable. Finally, digitoxin treatment, which inhibits Y576/577 FAK phosphorylation, abolished sexual dimorphism in HUVEC migration. These findings unravel complementary modulation of HUVEC functional phenotypes and signaling molecules involved in angiogenesis by hormone microenvironment and sex-specific factors, and highlight the need for sex-oriented pharmacological targeting of endothelial function.
Collapse
Affiliation(s)
- Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Chiara Baggio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Chiara Scapin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
19
|
Ma X, Zhou Y, Qiao B, Jiang S, Shen Q, Han Y, Liu A, Chen X, Wei L, Zhou L, Zhao J. Androgen aggravates liver fibrosis by activation of NLRP3 inflammasome in CCl 4-induced liver injury mouse model. Am J Physiol Endocrinol Metab 2020; 318:E817-E829. [PMID: 32182125 DOI: 10.1152/ajpendo.00427.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies have shown that there are differences between the sexes regarding to the occurrence and development of liver diseases, which may be associated with sex hormones. However, the mechanisms behind it are largely unknown. In this study, we first investigated the differences of liver injury between male and female mice, using the CCl4-induced liver injury mouse model. It showed that the liver damage of male mice was much more severe than that of female mice. Both the acute injury and fibrosis of the liver were reduced when androgens were depleted by castration of male mice. The vulnerability of male liver was associated with testis endocrine and excessive activation of inflammatory response in the liver. Castrated male mice with testosterone supplementation showed aggravated liver inflammatory response and fibrosis. The activity of NOD-like receptor protein 3 (NLRP3) inflammasome was increased when testosterone supplementation was provided. However, the enhanced inflammatory response and fibrosis due to testosterone supplementation were negated by inhibiting the activation of NLRP3 using the specific small molecule inhibitor MCC950. It suggests that testosterone is a key factor that influences liver injury by regulating the NLRP3 inflammasome activation-mediated inflammatory response.
Collapse
Affiliation(s)
- Xingyu Ma
- College of Animal Science, Southwest University, Chongqing, China
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yang Zhou
- College of Animal Science, Southwest University, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
- Laboratory Animal Center in College of Animal Science, Southwest University, Chongqing, China
| | - Bingke Qiao
- College of Animal Science, Southwest University, Chongqing, China
| | - Songhong Jiang
- College of Animal Science, Southwest University, Chongqing, China
| | - Qian Shen
- Department of Microbiology, Ohio State University, Columbus, Ohio
| | - Yuzhu Han
- College of Animal Science, Southwest University, Chongqing, China
| | - Anfang Liu
- College of Animal Science, Southwest University, Chongqing, China
| | - Xuequn Chen
- College of Animal Science, Southwest University, Chongqing, China
| | - Leiting Wei
- College of Animal Science, Southwest University, Chongqing, China
| | - Le Zhou
- College of Animal Science, Southwest University, Chongqing, China
| | - Jianjun Zhao
- College of Animal Science, Southwest University, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
- Laboratory Animal Center in College of Animal Science, Southwest University, Chongqing, China
| |
Collapse
|
20
|
Abstract
PURPOSE To determine whether testosterone supplementation is associated with retinal artery occlusion (RAO) or retinal vein occlusion (RVO). METHODS Retrospective matched cohort study using data from a large national U.S. insurance database. The testosterone cohort consisted of all male patients who filled a prescription for testosterone from 2000 to 2013. Five controls were matched on age (±3 years), sex, race, and similar time in plan (±3 months) for every exposed patient. Exclusion occurred for <2 years in the plan, <1 eye care visit, medications known to affect androgen levels, and systemic diseases associated with occlusions or increased testosterone. Cox proportional hazard regression assessed the hazard of a new diagnosis of RAO or RVO while controlling for age, race, diabetes mellitus, and hypertension. RESULTS A total of 35,784 incident testosterone users were compared with 178,860 matched controls. Ninety-three (0.3%) RAOs and 50 (0.1%) RVOs were found in the testosterone cohort and contrasted with 316 (0.2%) RAOs and 232 (0.1%) RVOs in the control group. After multivariate analysis, testosterone supplementation significantly increased the hazard of RAO (hazard ratio: 1.43, 95% confidence interval: 1.12-1.81, P = 0.004), but not of RVO (hazard ratio: 1.03, 95% confidence interval: 0.74-1.42, P = 0.86). CONCLUSION Although the incidence of RAO and RVO is low in users of testosterone, supplementation therapy is associated with an increased hazard of RAO, but apparently not of RVO.
Collapse
|
21
|
Ray PR, Khan J, Wangzhou A, Tavares-Ferreira D, Akopian AN, Dussor G, Price TJ. Transcriptome Analysis of the Human Tibial Nerve Identifies Sexually Dimorphic Expression of Genes Involved in Pain, Inflammation, and Neuro-Immunity. Front Mol Neurosci 2019; 12:37. [PMID: 30890918 PMCID: PMC6412153 DOI: 10.3389/fnmol.2019.00037] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/31/2019] [Indexed: 12/21/2022] Open
Abstract
Sex differences in gene expression are important contributors to normal physiology and mechanisms of disease. This is increasingly apparent in understanding and potentially treating chronic pain where molecular mechanisms driving sex differences in neuronal plasticity are giving new insight into why certain chronic pain disorders preferentially affect women vs. men. Large transcriptomic resources are now available and can be used to mine for sex differences to gather insight from molecular profiles using donor cohorts. We performed in-depth analysis of 248 human tibial nerve (hTN) transcriptomes from the GTEx Consortium project to gain insight into sex-dependent gene expression in the peripheral nervous system (PNS). We discover 149 genes with sex differential gene expression. Many of the more abundant genes in men are associated with inflammation and appear to be primarily expressed by glia or immune cells, with some genes downstream of Notch signaling. In women, we find the differentially expressed transcription factor SP4 that is known to drive a regulatory program, and may impact sex differences in PNS physiology. Many of these 149 differentially expressed (DE) genes have some previous association with chronic pain but few of them have been explored thoroughly. Additionally, using clinical data in the GTEx database, we identify a subset of DE, sexually dimorphic genes in diseases associated with chronic pain: arthritis and Type II diabetes. Our work creates a unique resource that identifies sexually dimorphic gene expression in the human PNS with implications for discovery of sex-specific pain mechanisms.
Collapse
Affiliation(s)
- Pradipta R. Ray
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Jawad Khan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Diana Tavares-Ferreira
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Armen N. Akopian
- Department of Endodontics, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
22
|
Gaba A, Mairhofer M, Zhegu Z, Leditznig N, Szabo L, Tschugguel W, Schneeberger C, Yotova I. Testosterone induced downregulation of migration and proliferation in human Umbilical Vein Endothelial Cells by Androgen Receptor dependent and independent mechanisms. Mol Cell Endocrinol 2018; 476:173-184. [PMID: 29777728 DOI: 10.1016/j.mce.2018.05.007] [Citation(s) in RCA: 5] [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] [Received: 08/30/2017] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022]
Abstract
Recent research has emphasized the potential unfavorable effects of declining testosterone (T) levels in men and the putative beneficial effect of androgen therapy in select women. Some controversy surrounding the mechanism of action and the effects of T on endothelium remains. In this study, we evaluated the mechanism of T action on pooled primary Human Umbilical Vein Endothelial Cells (HUVEC) of mixed gender by focusing on two important processes, proliferation and migration. In our in vitro model system, we found that only the supra-physiological dose of T affected these two processes irrespective of the ratio of male to female cells in the pools. At a concentration of 1 μM, T downregulated the proliferation of HUVEC by inducing arrest in the G1 cell cycle phase in an Androgen Receptor (AR)-independent manner. We show that treatment with 1 μM T also induced downregulation of HUVEC migration. This process was AR-dependent and was associated with persistent phosphorylation of ezrin, radixin and moesin. Regardless of the mechanism of action, the treatment of HUVEC with both supra- and physiological doses of T was associated with posttranscriptional stabilization of the AR upon ligand binding.
Collapse
Affiliation(s)
- Aulona Gaba
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | | | - Zyhdi Zhegu
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Nadja Leditznig
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Ladislaus Szabo
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Walter Tschugguel
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Christian Schneeberger
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Iveta Yotova
- Department of Gynecological Endocrinology, University Clinic of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
23
|
Sharif R, Bak-Nielsen S, Hjortdal J, Karamichos D. Pathogenesis of Keratoconus: The intriguing therapeutic potential of Prolactin-inducible protein. Prog Retin Eye Res 2018; 67:150-167. [PMID: 29758268 PMCID: PMC6235698 DOI: 10.1016/j.preteyeres.2018.05.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/25/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022]
Abstract
Keratoconus (KC) is the most common ectatic corneal disease, with clinical findings that include discomfort, visual disturbance and possible blindness if left untreated. KC affects approximately 1:400 to 1:2000 people worldwide, including both males and females. The aetiology and onset of KC remains a puzzle and as a result, the ability to treat or reverse the disease is hampered. Sex hormones are known to play a role in the maintenance of the structure and integrity of the human cornea. Hormone levels have been reported to alter corneal thickness, curvature, and sensitivity during different times of menstrual cycle. Surprisingly, the role of sex hormones in corneal diseases and KC has been largely neglected. Prolactin-induced protein, known to be regulated by sex hormones, is a new KC biomarker that has been recently proposed. Studies herein discuss the role of sex hormones as a control mechanism for KC onset and progression and evidence supporting the view that prolactin-induced protein is an important hormonally regulated biomarker in KC is discussed.
Collapse
Affiliation(s)
- Rabab Sharif
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, United States
| | - Sashia Bak-Nielsen
- Department of Ophthalmology, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Jesper Hjortdal
- Department of Ophthalmology, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Dimitrios Karamichos
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK 73104, United States; Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Science Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, United States.
| |
Collapse
|
24
|
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: 5.8] [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
|
25
|
Huxley VH, Kemp SS, Schramm C, Sieveking S, Bingaman S, Yu Y, Zaniletti I, Stockard K, Wang J. Sex differences influencing micro- and macrovascular endothelial phenotype in vitro. J Physiol 2018; 596:3929-3949. [PMID: 29885204 DOI: 10.1113/jp276048] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Endothelial dysfunction is an early hallmark of multiple disease states that also display sex differences with respect to age of onset, frequency and severity. Results of in vivo studies of basal and stimulated microvascular barrier function revealed sex differences that are difficult to ascribe to specific cells or environmental factors. The present study evaluated endothelial cells (EC) isolated from macro- and/or microvessels of reproductively mature rats under the controlled conditions of low-passage culture aiming to test the assumption that EC phenotype would be sex independent. The primary finding was that EC, regardless of where they are derived, retain a sex-bias in low-passage culture, independent of varying levels of reproductive hormones. The implications of the present study include the fallacy of expecting a universal set of mechanisms derived from study of EC from one sex and/or one vascular origin to apply uniformly to all EC under unstimulated conditions, and no less in disease. ABSTRACT Vascular endothelial cells (EC) are heterogeneous with respect to phenotype, reflecting at least the organ of origin, location within the vascular network and physical forces. As an independent influence on EC functions in health or aetiology, susceptibility, and progression of dysfunction in numerous disease states, sex has been largely ignored. The present study focussed on EC isolated from aorta (macrovascular) and skeletal muscle vessels (microvascular) of age-matched male and female rats under identical conditions of short-term (passage 4) culture. We tested the hypothesis that genomic sex would not influence endothelial growth, wound healing, morphology, lactate production, or messenger RNA and protein expression of key proteins (sex hormone receptors for androgen and oestrogens α and β; platelet endothelial cell adhesion molecule-1 and vascular endothelial cadherin mediating barrier function; αv β3 and N-cadherin influencing matrix interactions; intracellular adhesion molecule-1 and vascular cell adhesion molecule-1 mediating EC/white cell adhesion). The hypothesis was rejected because the EC origin (macro- vs. microvessel) and sex influenced multiple phenotypic characteristics. Statistical model analysis of EC growth demonstrated an hierarchy of variable importance, recapitulated for other phenotypic characteristics, with predictions assuming EC homogeneity < sex < vessel origin < sex and vessel origin. Furthermore, patterns of EC mRNA expression by vessel origin and by sex did not predict protein expression. Overall, the present study demonstrated that accurate assessment of sex-linked EC dysfunction first requires an understanding of EC function by position in the vascular tree and by sex. The results from a single EC tissue source/species/sex cannot provide universal insight into the mechanisms regulating in vivo endothelial function in health, and no less in disease.
Collapse
Affiliation(s)
- Virginia H Huxley
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Scott S Kemp
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Christine Schramm
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Steve Sieveking
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Susan Bingaman
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Yang Yu
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Isabella Zaniletti
- Department of Statistics, University of Missouri-Columbia, Columbia, MO, USA
| | - Kevin Stockard
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Jianjie Wang
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Biomedical Sciences, Missouri State University, Springfield, MO, USA
| |
Collapse
|
26
|
do Imperio GE, Bloise E, Javam M, Lye P, Constantinof A, Dunk C, Dos Reis FM, Lye SJ, Gibb W, Ortiga-Carvalho TM, Matthews SG. Chorioamnionitis Induces a Specific Signature of Placental ABC Transporters Associated with an Increase of miR-331-5p in the Human Preterm Placenta. Cell Physiol Biochem 2018; 45:591-604. [PMID: 29402780 PMCID: PMC7202864 DOI: 10.1159/000487100] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/10/2017] [Indexed: 12/16/2022] Open
Abstract
Background/Aims The ATP-binding cassette (ABC) transporters mediate drug biodisposition and immunological responses in the placental barrier. In vitro infective challenges alter expression of specific placental ABC transporters. We hypothesized that chorioamnionitis induces a distinct pattern of ABC transporter expression. Methods Gene expression of 50 ABC transporters was assessed using TaqMan® Human ABC Transporter Array, in preterm human placentas without (PTD; n=6) or with histological chorioamnionitis (PTDC; n=6). Validation was performed using qPCR, immunohistochemistry and Western blot. MicroRNAs known to regulate P-glycoprotein (P-gp) were examined by qPCR. Results Up-regulation of ABCB9, ABCC2 and ABCF2 mRNA was detected in chorioamnionitis (p<0.05), whereas placental ABCB1 (P-gp; p=0.051) and ABCG2 (breast cancer resistance protein-BCRP) mRNA levels (p=0.055) approached near significant up-regulation. In most cases, the magnitude of the effect significantly correlated to the severity of inflammation. Upon validation, increased placental ABCB1 and ABCG2 mRNA levels (p<0.05) were observed. At the level of immunohistochemistry, while BCRP was increased (p<0.05), P-gp staining intensity was significantly decreased (p<0.05) in PTDC. miR-331-5p, involved in P-gp suppression, was upregulated in PTDC (p<0.01) and correlated to the grade of chorioamnionitis (p<0.01). Conclusions Alterations in the expression of ABC transporters will likely lead to modified transport of clinically relevant compounds at the inflamed placenta. A better understanding of the potential role of these transporters in the events surrounding PTD may also enable new strategies to be developed for prevention and treatment of PTD.
Collapse
Affiliation(s)
- Guinever Eustaquio do Imperio
- Departments of Physiology, Toronto, Ontario, Canada.,Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Enrrico Bloise
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Departments of Morphology, Belo Horizonte, Brazil
| | - Mohsen Javam
- Departments of Physiology, Toronto, Ontario, Canada
| | | | | | - Caroline Dunk
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Stephen James Lye
- Departments of Physiology, Toronto, Ontario, Canada.,Obstetrics and Gynecology, Toronto, Ontario, Canada.,Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - William Gibb
- Departments of Obstetrics & Gynecology and Department of Cellular & Molecular Medicine, University of Ottawa, Toronto, Ontario, Canada
| | - Tania M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stephen Giles Matthews
- Departments of Physiology, Toronto, Ontario, Canada.,Obstetrics and Gynecology, Toronto, Ontario, Canada.,Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Differential sex-specific effects of oxygen toxicity in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2017; 486:431-437. [PMID: 28315681 DOI: 10.1016/j.bbrc.2017.03.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/14/2017] [Indexed: 11/21/2022]
Abstract
Despite the well-established sex-specific differences in the incidence of bronchopulmonary dysplasia (BPD), the molecular mechanism(s) behind these are not completely understood. Pulmonary angiogenesis is critical for alveolarization and arrest in vascular development adversely affects lung development. Human neonatal umbilical vein endothelial cells (HUVECs) provide a robust in vitro model for the study of endothelial cell physiology and function. Male and Female HUVECs were exposed to room air (21% O2, 5% CO2) or hyperoxia (95% O2, 5% CO2) for up to 72 h. Cell viability, proliferation, H2O2 production and angiogenesis were analyzed. Sex-specific differences in the expression of VEGFR2 and modulation of NF-kappa B pathway were measured. Male HUVECs have decreased survival, greater oxidative stress and impairment in angiogenesis compared to similarly exposed female cells. There is differential expression of VEGFR2 between male and female HUVECs and greater activation of the NF-kappa B pathway in female HUVECs under hyperoxic conditions. The results indicate that sex differences exist between male and female HUVECs in vitro after hyperoxia exposure. Since endothelial dysfunction has a major role in the pathogenesis of BPD, these differences could explain in part the mechanisms behind sex-specific differences in the incidence of this disease.
Collapse
|
28
|
Mudrovcic N, Arefin S, Van Craenenbroeck AH, Kublickiene K. Endothelial maintenance in health and disease: Importance of sex differences. Pharmacol Res 2017; 119:48-60. [PMID: 28108363 DOI: 10.1016/j.phrs.2017.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 02/07/2023]
Abstract
The vascular endothelium has emerged as more than just an inert monolayer of cells lining the vascular bed. It represents the interface between the blood stream and vessel wall, and has a strategic role in regulating vascular homeostasis by the release of vasoactive substances. Endothelial dysfunction contributes to the development and progression of cardiovascular disease. Recognition of sex-specific factors implicated in endothelial cell biology is important for the identification of clinically relevant preventive and/or therapeutic strategies. This review aims to give an overview of the recent advances in understanding the importance of sex specific observations in endothelial maintenance, both in healthy and diseased conditions. The female endothelium is highlighted in the context of polycystic ovary syndrome and pre-eclampsia. Furthermore, sex differences are explored in chronic kidney disease, which is currently appreciated as one of public health priorities. Overall, this review endorses integration of sex analysis in experimental and patient-oriented research in the exciting field of vascular biology.
Collapse
Affiliation(s)
- Neja Mudrovcic
- Department of Clinical Science, Intervention & Technology, Division of Obstetrics & Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Samsul Arefin
- Department of Clinical Science, Intervention & Technology, Division of Obstetrics & Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Amaryllis H Van Craenenbroeck
- Department of Nephrology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium; Department of Clinical Science, Intervention & Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Department of Clinical Science, Intervention & Technology, Division of Obstetrics & Gynecology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Science, Intervention & Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Gender Medicine, Department of Medicine-Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
29
|
Franconi F, Rosano G, Basili S, Montella A, Campesi I. Human cells involved in atherosclerosis have a sex. Int J Cardiol 2016; 228:983-1001. [PMID: 27915217 DOI: 10.1016/j.ijcard.2016.11.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/06/2016] [Indexed: 12/30/2022]
Abstract
The influence of sex has been largely described in cardiovascular diseases. Atherosclerosis is a complex process that involves many cell types such as vessel cells, immune cells and endothelial progenitor cells; however, many, if not all, studies do not report the sex of the cells. This review focuses on sex differences in human cells involved in the atherosclerotic process, emphasizing the role of sex hormones. Furthermore, we report sex differences and issues related to the processes that determine the fate of the cells such as apoptotic and autophagic mechanisms. The analysis of the data reveals that there are still many gaps in our knowledge regarding sex influences in atherosclerosis, largely for the cell types that have not been well studied, stressing the urgent need for a clear definition of experimental conditions and the inclusion of both sexes in preclinical studies.
Collapse
Affiliation(s)
- Flavia Franconi
- Assessorato alle Politiche per la Persona of Basilicata Region, Potenza, Italy; Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Rosano
- Cardiovascular and Cell Sciences Research Institute, St. George's University of London, United Kingdom
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties - Research Center on Gender and Evaluation and Promotion of Quality in Medicine (CEQUAM), Sapienza University of Rome, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Osilo, Italy.
| |
Collapse
|
30
|
Androgen actions on endothelium functions and cardiovascular diseases. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 13:183-96. [PMID: 27168746 PMCID: PMC4854959 DOI: 10.11909/j.issn.1671-5411.2016.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
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
|
31
|
Torres-Estay V, Carreño DV, San Francisco IF, Sotomayor P, Godoy AS, Smith GJ. Androgen receptor in human endothelial cells. J Endocrinol 2015; 224:R131-7. [PMID: 25563353 PMCID: PMC4700832 DOI: 10.1530/joe-14-0611] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Androgen receptor (AR) is a ligand-inducible transcription factor, and a member of the steroid-thyroid-retinoid receptor superfamily, that mediates the biological effects of androgens in a wide range of physiological and pathological processes. AR expression was identified in vascular cells nearly 20 years ago, and recent research has shown that AR mediates a variety of actions of androgens in endothelial and vascular smooth muscle cells. In this mini-review, we review evidence indicating the importance of AR in human endothelial cell (HUVEC) homeostatic and pathogenic processes. Although a role for AR in the modulation of HUVEC biology is evident, the molecular mechanisms by which AR regulates HUVEC homeostasis and disease processes are not fully understood. Understanding these mechanisms could provide critical insights into the processes of pathogenesis of diseases ranging from cardiovascular disease to cancer that are major causes of human morbidity and mortality.
Collapse
Affiliation(s)
- Verónica Torres-Estay
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - Daniela V Carreño
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - Ignacio F San Francisco
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - Paula Sotomayor
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - Alejandro S Godoy
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | - Gary J Smith
- Departamento de FisiologíaUrologíaPontificia Universidad Católica de Chile, Santiago de Chile, ChileCenter for Integrative Medicine and Innovative SciencesUniversidad Andrés Bello, Santiago de Chile, ChileDepartment of UrologyRoswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| |
Collapse
|
32
|
Imperlini E, Mancini A, Alfieri A, Martone D, Caterino M, Orrù S, Buono P. Molecular effects of supraphysiological doses of doping agents on health. MOLECULAR BIOSYSTEMS 2015; 11:1494-506. [DOI: 10.1039/c5mb00030k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Supraphysiological doses of doping agents, such as T/DHT and GH/IGF-1, affect cellular pathways associated with apoptosis and inflammation.
Collapse
Affiliation(s)
| | - Annamaria Mancini
- Dipartimento di Scienze Motorie e del Benessere
- Università “Parthenope” di Napoli
- 80133 Naples
- Italy
- CEINGE Biotecnologie Avanzate s.c. a r.l
| | - Andreina Alfieri
- Dipartimento di Scienze Motorie e del Benessere
- Università “Parthenope” di Napoli
- 80133 Naples
- Italy
- CEINGE Biotecnologie Avanzate s.c. a r.l
| | - Domenico Martone
- Dipartimento di Scienze Motorie e del Benessere
- Università “Parthenope” di Napoli
- 80133 Naples
- Italy
| | | | - Stefania Orrù
- Dipartimento di Scienze Motorie e del Benessere
- Università “Parthenope” di Napoli
- 80133 Naples
- Italy
- CEINGE Biotecnologie Avanzate s.c. a r.l
| | - Pasqualina Buono
- IRCCS SDN
- Naples
- Italy
- Dipartimento di Scienze Motorie e del Benessere
- Università “Parthenope” di Napoli
| |
Collapse
|
33
|
Addis R, Campesi I, Fois M, Capobianco G, Dessole S, Fenu G, Montella A, Cattaneo MG, Vicentini LM, Franconi F. Human umbilical endothelial cells (HUVECs) have a sex: characterisation of the phenotype of male and female cells. Biol Sex Differ 2014; 5:18. [PMID: 25535548 PMCID: PMC4273493 DOI: 10.1186/s13293-014-0018-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/20/2014] [Indexed: 01/04/2023] Open
Abstract
Background Human umbilical endothelial cells (HUVECs) are widely used to study the endothelial physiology and pathology that might be involved in sex and gender differences detected at the cardiovascular level. This study evaluated whether HUVECs are sexually dimorphic in their morphological, proliferative and migratory properties and in the gene and protein expression of oestrogen and androgen receptors and nitric oxide synthase 3 (NOS3). Moreover, because autophagy is influenced by sex, its degree was analysed in male and female HUVECs (MHUVECs and FHUVECs). Methods Umbilical cords from healthy, normal weight male and female neonates born to healthy non-obese and non-smoking women were studied. HUVEC morphology was analysed by electron microscopy, and their function was investigated by proliferation, viability, wound healing and chemotaxis assays. Gene and protein expression for oestrogen and androgen receptors and for NOS3 were evaluated by real-time PCR and Western blotting, respectively, and the expression of the primary molecules involved in autophagy regulation [protein kinase B (Akt), mammalian target of rapamycin (mTOR), beclin-1 and microtubule-associated protein 1 light chain 3 (LC3)] were detected by Western blotting. Results Cell proliferation, migration NOS3 mRNA and protein expression were significantly higher in FHUVECs than in MHUVECs. Conversely, beclin-1 and the LC3-II/LC3-I ratio were higher in MHUVECs than in FHUVECs, indicating that male cells are more autophagic than female cells. The expression of oestrogen and androgen receptor genes and proteins, the protein expression of Akt and mTOR and cellular size and shape were not influenced by sex. Body weights of male and female neonates were not significantly different, but the weight of male babies positively correlated with the weight of the mother, suggesting that the mother’s weight may exert a different influence on male and female babies. Conclusions The results indicate that sex differences exist in prenatal life and are parameter-specific, suggesting that HUVECs of both sexes should be used as an in vitro model to increase the quality and the translational value of research. The sex differences observed in HUVECs could be relevant in explaining the diseases of adulthood because endothelial dysfunction has a crucial role in the pathogenesis of cardiovascular diseases, diabetes mellitus, neurodegeneration and immune disease.
Collapse
Affiliation(s)
- Roberta Addis
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari Italy
| | - Marco Fois
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Surgical, Microsurgical and Medical Sciences, Gynaecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Salvatore Dessole
- Department of Surgical, Microsurgical and Medical Sciences, Gynaecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Grazia Fenu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - Lucia M Vicentini
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano, Italy
| | - Flavia Franconi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari Italy.,Assessorato alle Politiche per la Persona, Region Basilicata, Italy
| |
Collapse
|
34
|
Medenwald D, Girndt M, Loppnow H, Kluttig A, Nuding S, Tiller D, Thiery JJ, Greiser KH, Haerting J, Werdan K. Inflammation and renal function after a four-year follow-up in subjects with unimpaired glomerular filtration rate: results from the observational, population-based CARLA cohort. PLoS One 2014; 9:e108427. [PMID: 25259714 PMCID: PMC4178159 DOI: 10.1371/journal.pone.0108427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/20/2014] [Indexed: 12/03/2022] Open
Abstract
Background There is evidence that chronic inflammation is associated with the progression/development of chronic renal failure; however, relations in subjects with preserved renal function remain insufficiently understood. Objective To examine the association of inflammation with the development of renal failure in a cohort of the elderly general population. Methods After excluding subjects with reduced estimated glomerular filtration rate (eGFR<60 mL/min/1.73 m2) and missing data, the cohort incorporated 785 men and 659 women (aged 45–83 years). Follow-up was performed four years after baseline. Covariate adjusted linear and logistic regression models were used to assess the association of plasma/serum concentrations of soluble tumour necrosis factor receptor 1 (sTNF-R1), C-reactive protein (CRP), and interleukin 6 (IL-6) with change in eGFR/creatinine. The areas under the curve (AUCs) from receiver operating characteristics (ROCs) were estimated. Results In adjusted models sTNF-R1 was distinctively associated with a decline in eGFR in men (0.6 mL/min/1.73 m2 per 100 pg/mL sTNF-R1; 95% CI: 0.4–0.8), but not in women. A similar association could not be found for CRP or IL-6. Estimates of sTNF-R1 in the cross-sectional analyses were similar between sexes, while CRP and IL-6 were not relevantly associated with eGFR/creatinine. Conclusion In the elderly male general population with preserved renal function sTNF-R1 predicts the development of renal failure.
Collapse
Affiliation(s)
- Daniel Medenwald
- Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
- * E-mail:
| | - Matthias Girndt
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Harald Loppnow
- Department of Internal Medicine III, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Alexander Kluttig
- Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Sebastian Nuding
- Department of Internal Medicine III, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Daniel Tiller
- Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Joachim J. Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Karin H. Greiser
- German Cancer Research Centre, Division of Cancer Epidemiology, Heidelberg, Germany
| | - Johannes Haerting
- Institute of Medical Epidemiology, Biostatistics and Informatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Karl Werdan
- Department of Internal Medicine III, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|