1
|
AbuMadighem A, Rubin E, Arazi E, Lunenfeld E, Huleihel M. Adrenocorticotropic hormone and its receptor as a novel testicular system involves in the development of spermatogenesis. Life Sci 2025; 368:123480. [PMID: 39978588 DOI: 10.1016/j.lfs.2025.123480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/17/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
AIMS To identify functional membrane-associate-specific SSC markers and examine the development of these cells under in vitro conditions. MATERIALS AND METHODS Cells were enzymatically isolated from seminiferous tubules (STs) of immature mice. Spermatogonial cells (Thy1, alpha-6-integrin, and C-KIT) were sorted by FACS. RNA was extracted from these cells for RNAseq analysis. The effect of adrenocorticotropic hormone (ACTH) - the ligand of MC2R- on the development of mouse spermatogonial cells was performed in vitro using a methylcellulose culture system (MCS). Immunofluorescence staining was used to localize MC2R-positive cells in the testes of immature and adult humans and mice and testes of busulfan-treated immature mice. KEY FINDINGS Our RNAseq analysis revealed a high expression of melanocortin receptor 2 (MC2R) in Thy1-positive sorted cells. MC2R-positive cells were localized in the periphery of the STs of humans (prepubertal and adults) and mice at immature and adult ages (normal and busulfan-treated mice). MC2R was doubled stained with PLZF and CDH1 (SSC markers). ACTH was localized in mouse testicular germ cells (pre-meiotic, meiotic, and post-meiotic cells) and somatic cells (Sertoli, Leydig, and peritubular cells). The addition of ACTH to isolated cells from mouse STs in MCS significantly increased the development of pre-meiotic and meiotic/post-meiotic cells in vitro. SIGNIFICANCE We were able to identify, for the first time, a novel membrane-associated and functional SSC marker (MC2R) with relation to ACTH. This marker can be used in future male fertility preservation strategies. Furthermore, we explored a novel testicular system (ACTH system) that regulates the development of spermatogenesis.
Collapse
Affiliation(s)
- Ali AbuMadighem
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eitan Rubin
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eden Arazi
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Mahmoud Huleihel
- The Shraga Segal Dept. Microbiology, Immunology and Genetics, Israel; The Center of Advanced Research and Education in Reproduction (CARER), Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
2
|
Kyrgiafini MA, Kaltsas A, Chatziparasidou A, Mamuris Z. The Small RNA Landscape in Azoospermia: Implications for Male Infertility and Sperm Retrieval-A Preliminary Study. Int J Mol Sci 2025; 26:3537. [PMID: 40331996 PMCID: PMC12027063 DOI: 10.3390/ijms26083537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
MicroRNAs (miRNAs), a class of small noncoding RNAs, play a crucial role in spermatogenesis. However, their specific expression patterns in azoospermic patients, particularly in relation to sperm presence and pregnancy outcomes, remain underexplored. We performed small RNA sequencing on forty testicular tissue samples from idiopathic azoospermic and cryptozoospermic patients who underwent testicular sperm extraction (TESE). Differentially expressed (DE) miRNAs were identified across groups with high, rare, or no spermatozoa presence, as well as between individuals with successful and unsuccessful pregnancies following assisted reproduction. Functional enrichment analyses were conducted to assess the biological relevance of miRNA alterations. Our findings revealed distinct miRNA expression patterns linked to sperm presence and pregnancy outcomes. Samples with high sperm presence exhibited reduced miRNA expression, while those with impaired spermatogenesis demonstrated upregulated miRNAs associated with cell survival and differentiation pathways. Several regulatory pathways were also disrupted in samples leading to unsuccessful pregnancies, including the estrogen signaling receptor (ESR) pathway, interleukin-4 and interleukin-13 signaling, and transcription networks. This study highlights miRNA-mediated regulatory differences in azoospermic patients, identifying potential biomarkers for sperm retrieval success and fertility outcomes. Future validation and multi-omics approaches are needed to confirm these findings and enhance male infertility diagnostics.
Collapse
Affiliation(s)
- Maria-Anna Kyrgiafini
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| | - Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Alexia Chatziparasidou
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
- Embryolab IVF Unit, St. 173-175 Ethnikis Antistaseos, Kalamaria, 55134 Thessaloniki, Greece
| | - Zissis Mamuris
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|
3
|
Chen J, Guo JM, Jiang BJ, Sun FY, Qu YC. Impact of physical activity on semen quality: a review of current evidence. Asian J Androl 2025:00129336-990000000-00294. [PMID: 40084407 DOI: 10.4103/aja20252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 03/16/2025] Open
Abstract
A growing global trend indicates a decline in semen quality, with a lack of physical activity identified as one of the contributing factors. Exercise is medication, and numerous studies have explored its effects on semen quality. However, there is no consensus on the most effective type and intensity of exercise for improving semen quality, owing to inconsistent findings across studies. These discrepancies may be attributable to variations in study populations (e.g., healthy versus infertile individuals) and research methodologies (e.g., observational versus interventional studies). This paper reviews the existing literature from the databases PubMed, Web of Science, and Google Scholar, reclassifying articles on their subject and research designs to delineate the relationship between exercise and semen quality. It also summarizes the mechanisms through which exercise influences semen quality, including hormonal regulation, oxidative stress, and inflammatory factors.
Collapse
Affiliation(s)
- Jing Chen
- Institute of Artificial Intelligence in Sports (IAIS), Capital University of Physical and Sports, Beijing 100000, China
| | | | | | | | | |
Collapse
|
4
|
Aspesi D, Bass N, Kavaliers M, Choleris E. The Role of Androgens and Estrogens in Social Interactions and Social Cognition. Neuroscience 2025; 568:476-502. [PMID: 37080448 DOI: 10.1016/j.neuroscience.2023.03.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 03/02/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Gonadal hormones are becoming increasingly recognized for their effects on cognition. Estrogens, in particular, have received attention for their effects on learning and memory that rely upon the functioning of various brain regions. However, the impacts of androgens on cognition are relatively under investigated. Testosterone, as well as estrogens, have been shown to play a role in the modulation of different aspects of social cognition. This review explores the impact of testosterone and other androgens on various facets of social cognition including social recognition, social learning, social approach/avoidance, and aggression. We highlight the relevance of considering not only the actions of the most commonly studied steroids (i.e., testosterone, 17β-estradiol, and dihydrotestosterone), but also that of their metabolites and precursors, which interact with a plethora of different receptors and signalling molecules, ultimately modulating behaviour. We point out that it is also essential to investigate the effects of androgens, their precursors and metabolites in females, as prior studies have mostly focused on males. Overall, a comprehensive analysis of the impact of steroids such as androgens on behaviour is fundamental for a full understanding of the neural mechanisms underlying social cognition, including that of humans.
Collapse
Affiliation(s)
- Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Canada
| | - Noah Bass
- Department of Psychology and Neuroscience Program, University of Guelph, Canada
| | - Martin Kavaliers
- Department of Psychology and Neuroscience Program, University of Guelph, Canada; Department of Psychology, University of Western Ontario, London, Canada; Graduate Program in Neuroscience, University of Western Ontario, London, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Canada.
| |
Collapse
|
5
|
Pozzi E, Corsini C, Salonia A. Medical therapy for male infertility. Curr Opin Urol 2025; 35:157-164. [PMID: 39380445 DOI: 10.1097/mou.0000000000001231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
PURPOSE OF REVIEW To provide up-to-date evidence and clinical guidance on the role of medical therapy in the context of hormonal imbalances affecting human spermatogenesis. RECENT FINDINGS Compelling evidence has accumulated over the years regarding the role of gonadotropins, selective estrogen modulators, and aromatase inhibitors to either improve or restore spermatogenesis in men with hormonal abnormalities (e.g. hypogonadotropic/hypergonadotropic hypogonadism, hyperprolactinemia) or supraphysiologic levels (e.g. exogenous testosterone/anabolic steroid use). Despite the increasing number of studies being performed, most of the available evidence relies on small nonrandomized studies, mainly in men with hypergonadotropic hypogonadism or with history of exogenous testosterone/anabolic steroid use. As such, the efficacy of medical therapy is highly variable emphasizing the necessity of randomized clinical trials and individualized approaches. SUMMARY This narrative review provides clinical guidance on medical therapies for male factor infertility based on the most up-to-date evidence, focusing on treatments for hormonal abnormalities (either hypogonadotropic or hypergonadotropic hypogonadism and hyperprolactinemia) and supraphysiologic levels (and exogenous testosterone/anabolic steroid use) to improve spermatogenesis.
Collapse
Affiliation(s)
- Edoardo Pozzi
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Christian Corsini
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
6
|
Celik Atalay E, Er Demirhan B, Sagdıcoglu Celep AG. Low-Calorie Sweeteners and Reproductive Health: Evidence and Debates. CURRENT NUTRITION & FOOD SCIENCE 2025; 21:309-332. [DOI: 10.2174/0115734013315621240802055207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/02/2024] [Accepted: 06/21/2024] [Indexed: 01/04/2025]
Abstract
The reduction in sugar consumption has led to increased use of low-calorie artificial
sweeteners. This coincides with an increase in infertility rates, suggesting that low-calorie artificial
sweeteners may negatively affect reproductive health. Low-calorie sweeteners may affect
oxidative stress, glucose regulation, and the microbiota, which are associated with reproductive
health. Therefore, a review was conducted to examine the effects of commonly used low-calorie
sweeteners on reproductive health through potential biological mechanisms. This review addresses
the effects of low-calorie sweeteners in a wide range of areas, such as infertility, pregnancy and
neonatal health, and early menarche. Recent studies have indicated potential adverse effects of artificial
sweeteners on reproductive health. Research has examined the potential impacts of artificial
sweeteners on various parameters, such as hormone levels, sperm quality, sperm motility, ovarian
function, and pregnancy outcomes. However, the findings of current studies are inconsistent, and
these disparate results may stem from metabolic differences among different types of artificial
sweeteners, variations in research methodologies, diversity in sample sizes, and fluctuations in
study populations. Therefore, further research is needed to comprehensively understand the effects
of artificial sweeteners on reproductive health.
Collapse
Affiliation(s)
- Ece Celik Atalay
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Turkey
| | - Buket Er Demirhan
- Department of
Pharmaceutical Basic Science, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | | |
Collapse
|
7
|
Talebi SF, Seify M, Bhandari RK, Shoorei H, Oskuei SD. Fluoride-induced testicular and ovarian toxicity: evidence from animal studies. Biol Res 2025; 58:6. [PMID: 39863878 PMCID: PMC11762501 DOI: 10.1186/s40659-025-00586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Fluoride (F), as a natural element found in a wide range of sources such as water and certain foods, has been proven to be beneficial in preventing dental caries, but concerns have been raised regarding its potential deleterious effects on overall health. Sodium fluoride (NaF), another form of F, has the ability to accumulate in reproductive organs and interfere with hormonal regulation and oxidative stress pathways, contributing to reproductive toxicity. While the exact mechanisms of F-induced reproductive toxicity are not fully understood, this review aims to elucidate the mechanisms involved in testicular and ovarian injury. In males, F exposure at different doses has been associated with reduced testis weight, reduced sperm quality in terms of count, motility, and viability, as well as abnormal sperm morphology and disruption of seminiferous tubules by altering hormone levels (especially testosterone), impairing spermatogenesis, and inducing oxidative stress and zinc deficiency. Similarly, administration of F can impact female reproductive health by affecting ovarian function, hormone levels, oocyte quality, and the regularity of the estrous cycle. However, the impact of F exposure on LH, FSH, and GnRH levels is controversial between males and females. In both males and females, F exerts its adverse effects by triggering apoptosis, autophagy, inflammation, mitochondrial dysfunction, reduction in ATP synthesis, and modulation of important genes involved in steroidogenesis. Furthermore, genetic susceptibility and individual variations in F metabolism may contribute to different responses to fluoride exposure.
Collapse
Affiliation(s)
| | - Mohammad Seify
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ramji Kumar Bhandari
- Division of Biological Sciences, University of Missouri, Columbia, MO, 65211, USA.
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Shahram Dabiri Oskuei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Scandlan OLM, Favetta LA. Do Delta-9-tetrahydrocannabinol and Cannabidiol have opposed effects on male fertility? Toxicol Lett 2025; 403:94-104. [PMID: 39657895 DOI: 10.1016/j.toxlet.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Cannabis sativa is a complex plant, renowned for its diverse array of bioactive compounds, the most prominent of which are delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds exhibit markedly opposing pharmacological effects, with THC being primarily psychoactive and CBD known for its non-psychoactive properties. In recent years, there has been growing interest in the potential health implications of these compounds, particularly concerning male reproductive health. Accumulating evidence over the past decade has alluded to the potential negative effects of THC, including its association with reduced sperm quality, altered hormone levels, changes in genetic and epigenetic profiles, and potential impacts on fertility. Conversely, emerging studies suggest that CBD may exert protective and beneficial effects on male reproductive health, possibly through its anti-inflammatory and antioxidant properties. This review aims to provide a comprehensive analysis of the current scientific literature, delineating the mechanisms by which THC and CBD influence male reproductive health, highlighting the disparities in their effects, and discussing the clinical and therapeutic implications of these findings.
Collapse
Affiliation(s)
- Olivia L M Scandlan
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Laura A Favetta
- Reproductive Health and Biotechnology Laboratory, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
9
|
Pande A, Kinkade CW, Prout N, Chowdhury SF, Rivera-Núñez Z, Barrett ES. Prenatal exposure to synthetic chemicals in relation to HPA axis activity: A systematic review of the epidemiological literature. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 956:177300. [PMID: 39488279 DOI: 10.1016/j.scitotenv.2024.177300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Pregnant people are widely exposed to numerous synthetic chemicals with known endocrine-disrupting properties (e.g., phthalates, phenols, per- and poly-fluoroalkyl substances (PFAS)). To date, most epidemiological research on how endocrine-disrupting chemicals (EDCs) disrupt hormone pathways has focused on estrogens, androgens, and thyroid hormones. Far less research has examined the impact of EDCs on the hypothalamic-pituitary-adrenal (HPA) axis, despite its central role in the physiologic stress response and metabolic function. OBJECTIVE To systematically review the epidemiological literature on prenatal synthetic EDC exposures in relation to HPA axis hormones (e.g., corticotropin-releasing hormone, adrenocorticotropic hormone, cortisol, cortisone) in pregnant people and their offspring. METHODS A literature search of PubMed, Scopus, and Embase was conducted. Primary research studies were selected for inclusion by two independent reviewers and risk of bias was assessed using the Office of Health Assessment and Translation guidelines established by the National Toxicology Program with customization for the specific research topic. Data were extracted from each study and included in a qualitative synthesis. RESULTS 22 published studies met the inclusion criteria. Phthalates were the most prevalent EDC studied, followed by PFAS, phenols, and parabens, with fewer studies considering other synthetic chemicals. Offspring glucocorticoids were the most commonly considered outcome, followed by maternal glucocorticoids and placental corticotropin-releasing hormone. There was considerable heterogeneity in methods across studies, particularly in HPA axis outcome measures and matrices, making cross-study comparisons challenging. Numerous studies suggested disruption of HPA axis hormones and sex differences in association, but results varied considerably across studies and EDC classes. CONCLUSIONS The limited literature to date suggests the HPA axis may be vulnerable to disruption by synthetic EDCs. Carefully designed studies that prioritize biospecimen collection specific to HPA axis hormones are needed along with greater standardization of biospecimen collection and analysis protocols to facilitate cross-study comparisons and interpretation.
Collapse
Affiliation(s)
- Anushka Pande
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Carolyn W Kinkade
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Nashae Prout
- Wynne Center for Family Research, University of Rochester, Rochester, NY 14642, USA; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Sadia F Chowdhury
- Wynne Center for Family Research, University of Rochester, Rochester, NY 14642, USA; Translational Biomedical Sciences Program, University of Rochester, Rochester, NY 14642, USA
| | - Zorimar Rivera-Núñez
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854, USA
| | - Emily S Barrett
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854, USA.
| |
Collapse
|
10
|
Gómez-Olarte S, Mailänder V, Castro-Neves J, Stojanovska V, Schumacher A, Meyer N, Zenclussen AC. The ENDOMIX perspective: how everyday chemical mixtures impact human health and reproduction by targeting the immune system†. Biol Reprod 2024; 111:1170-1187. [PMID: 39446589 DOI: 10.1093/biolre/ioae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Endocrine-disrupting chemicals are natural and synthetic compounds found ubiquitously in the environment that interfere with the hormonal-immune axis, potentially impacting human health and reproduction. Exposure to endocrine-disrupting chemicals has been associated with numerous health risks, such as neurodevelopmental disorders, metabolic syndrome, thyroid dysfunction, infertility, and cancers. Nevertheless, the current approach to establishing causality between these substances and disease outcomes has limitations. Epidemiological and experimental research on endocrine-disrupting chemicals faces challenges in accurately assessing chemical exposure and interpreting non-monotonic dose response curves. In addition, most studies have focused on single chemicals or simple mixtures, overlooking complex real-life exposures and mechanistic insights, in particular regarding endocrine-disrupting chemicals' impact on the immune system. The ENDOMIX project, funded by the EU's Horizon Health Program, addresses these challenges by integrating epidemiological, risk assessment, and immunotoxicology methodologies. This systemic approach comprises the triangulation of human cohort, in vitro, and in vivo data to determine the combined effects of chemical mixtures. The present review presents and discusses current literature regarding human reproduction in the context of immunotolerance and chemical disruption mode of action. It further underscores the ENDOMIX perspective to elucidate the impact of endocrine-disrupting chemicals on immune-reproductive health.
Collapse
Affiliation(s)
- Sergio Gómez-Olarte
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Verena Mailänder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Júlia Castro-Neves
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Violeta Stojanovska
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Anne Schumacher
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Nicole Meyer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| |
Collapse
|
11
|
De Silva NL, Papanikolaou N, Grossmann M, Antonio L, Quinton R, Anawalt BD, Jayasena CN. Male hypogonadism: pathogenesis, diagnosis, and management. Lancet Diabetes Endocrinol 2024; 12:761-774. [PMID: 39159641 DOI: 10.1016/s2213-8587(24)00199-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 08/21/2024]
Abstract
Organic male hypogonadism due to irreversible hypothalamic-pituitary-testicular (HPT) pathology is easily diagnosed and treated with testosterone-replacement therapy. However, controversy surrounds the global practice of prescribing testosterone to symptomatic men with low testosterone and non-gonadal factors reducing health status, such as obesity, type 2 diabetes, and ageing (ie, functional hypogonadism), but without identifiable HPT axis pathology. Health optimisation remains the gold-standard management strategy. Nevertheless, in the last decade large clinical trials and an individual patient data meta-analysis of smaller clinical trials confirmed that testosterone therapy induces modest, yet statistically significant, improvements in sexual function without increasing short-term to medium-term cardiovascular or prostate cancer risks in men with functional hypogonadism. Although testosterone improves bone mineral density and insulin sensitivity in these men, trials from the last decade suggest insufficient evidence to determine the safety and effectiveness of use of this hormone for the prevention of fractures or type 2 diabetes. This Review discusses the pathogenesis and diagnosis of male hypogonadism and appraises the evidence underpinning the management of this condition.
Collapse
Affiliation(s)
- Nipun Lakshitha De Silva
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Faculty of Medicine, General Sir John Kotelawala Defence University, Colombo, Sri Lanka
| | - Nikoleta Papanikolaou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Mathis Grossmann
- Department of Medicine (Austin Health), The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology, Austin Health, Heidelberg, VIC, Australia
| | - Leen Antonio
- Department of Chronic Diseases and Metabolism (CHROMETA), Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium; Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Richard Quinton
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Northern Regional Gender Dysphoria Service, Cumbria Northumberland Tyne & Wear NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Bradley David Anawalt
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Channa N Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
12
|
Zheng XM, Zhang XD, Tan LL, Zhang J, Wang TT, Ling Q, Wang H, Ouyang KW, Wang KW, Chang W, Li H, Zhu HL, Xiong YW, Wang H. Sirt1 m6A modification-evoked Leydig cell senescence promotes Cd-induced testosterone decline. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116884. [PMID: 39153281 DOI: 10.1016/j.ecoenv.2024.116884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024]
Abstract
Diminished testosterone levels have been documented as a key factor in numerous male health disorders. Both human and animal studies have consistently demonstrated that cadmium (Cd), a pervasive environmental heavy metal, results in decreased testosterone levels. However, the exact mechanism through which Cd interferes with testosterone synthesis remains incompletely elucidated. This research sought to examine the impact of cellular senescence on Cd-suppressed testosterone synthesis. We also investigated the related m6A modification mechanism. The results demonstrated that Cd (100 mg/L) led to a decrease in testosterone levels, along with downregulated expression of testosterone synthase in C57BL/6 N male mice. Furthermore, Cd significantly increased β-galactosidase staining intensity, senescence-related proteins, and senescence-related secretory phenotypes in mouse testicular Leydig cells. Subsequent investigations revealed that Cd decreased the mRNA and protein levels of NAD-dependent deacetylase Sirtuin-1 (SIRT1) in Leydig cells. Mechanistically, mice treated with resveratrol (50 mg/kg), a specific SIRT1 activator, mitigated Leydig cell senescence and reversed Cd-reduced testosterone levels in mouse testes. These effects were also restored by SIRT1 overexpression in Leydig cells. Additionally, we found that Cd increased the level of methyltransferase enzyme METTL3 and Sirt1 m6A modification in Leydig cells. Mettl3 siRNA effectively restored Cd-enhanced Sirt1 m6A level and reversed Cd-downregulated Sirt1 mRNA expression in Leydig cells. Overall, our findings suggest that Cd exposure inhibits testosterone synthesis via Sirt1 m6A modification-mediated senescence in mouse testes. These results offer an experimental basis for investigating the causes and potential treatments of hypotestosteronemia induced by environmental factors.
Collapse
Affiliation(s)
- Xin-Mei Zheng
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Xu-Dong Zhang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Lu-Lu Tan
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Jin Zhang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Tian-Tian Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Qing Ling
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Kong-Wen Ouyang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Kai-Wen Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Wei Chang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hao Li
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China.
| | - Hua Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| |
Collapse
|
13
|
Sahoo P, Sarkar D, Sharma S, Verma A, Naik SK, Prashar V, Parkash J, Singh SK. Knockdown of type 2 orexin receptor in adult mouse testis potentiates testosterone production and germ cell proliferation. Mol Cell Endocrinol 2024; 592:112312. [PMID: 38866320 DOI: 10.1016/j.mce.2024.112312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Orexins (OXs) are neuropeptides which regulate various physiological processes. OXs exist in two different forms, mainly orexin A (OXA) and orexin B (OXB) and their effects are mediated via OX1R and OX2R. Presence of OXB and OX2R in mouse testis is also reported. However, the role of OXB/OX2R in the male gonad remains unexplored. Herein we investigated the role of OXB/OX2R system in testicular physiology under in vivo and ex vivo conditions. Adult mice were given a single dose of bilateral intratesticular injection of siRNA targeting OX2R and were sacrificed 96 h post-injection. OX2R-knockdown potentiated serum and intratesticular testosterone levels with up-regulation in the expressions of major steroidogenic proteins. Germ cell proliferation also increased in siRNA-treated mice. Results of the ex vivo experiment also supported the findings of the in vivo study. In conclusion, OX2R may regulate testosterone production and thereby control the fine-tuning between steroidogenesis and germ cell dynamics.
Collapse
Affiliation(s)
- Pratikshya Sahoo
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Debarshi Sarkar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India.
| | - Shubhangi Sharma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Arpit Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Suraj Kumar Naik
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO-Ghudda, Bathinda, 151401, India
| | - Shio Kumar Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| |
Collapse
|
14
|
Rodriguez-Sánchez AJ, Meza-Herrera CA, De Santiago-Miramontes A, Navarrete-Molina C, Veliz-Deras FG, Ordoñez-Morales JZ, Flores-Salas JM, Marin-Tinoco RI. Circular Economy, Dairy Cow Feed Leftovers, and Withania somnifera Supplementation: Effects on Black Belly Ram's Libido, Sperm Quality, Sexual Behavior, and Hemogram Values. BIOLOGY 2024; 13:656. [PMID: 39336084 PMCID: PMC11428648 DOI: 10.3390/biology13090656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024]
Abstract
Considering a circular economy perspective, this study evaluates the possible effect of targeted short-term supplementation with Withania somnifera L. (WS; Ashwagandha) on ram's seminal quality, socio-sexual behaviors, and blood constituents. Black Belly rams (n = 20) received a basal diet comprising feed-leftovers from dairy cows in the north-arid Mexico (i.e., Comarca Lagunera CL). The experimental units, with proven libido and fertility, were homogeneous in terms of age (3.41 ± 0.21 yr.), live weight (LW; 53.8 ± 3.3 kg), body condition (BC; 2.96 ± 0.01 units), initial sperm concentration (2387 ± 804 × 106), and viability (23.9 ± 15.6%). Rams were randomly assigned during the transition reproductive period (i.e., May to Jun; 25° NL) to three treatment groups: non-supplemented control group (CONT; n = 6), low WS-supplemented (LWS; i.e., 100 mg kg LW-1 d-1 × 40 d; n = 7), and high-WS-supplemented (HWS; i.e., 200 mg kg LW-1 d-1 × 40 d; n = 7). The basal leftover diet was offered twice daily (0700 and 1600 h); the experimental period (EP) lasted 47 d. No differences (p > 0.05) among treatments occurred regarding LW and BCS at the onset of the EP. Whereas the greater scrotal circumference (SCRC, cm) arose in the LWS and CONT rams, an increased ejaculated volume (VOLEJA, mL) occurred in the WS-rams. A total of 5/9 (i.e., 55%) appetitive and 3/3 (i.e., 100%) consummatory sexual behaviors favored (p < 0.05) the WS-rams, particularly the HWS rams, towards the final EP. The same was true (p < 0.05) regarding the hemogram variables white blood cell count (×109 cells L-1), hemoglobin concentration (g dL-1), and medium corpuscular volume (fL). This study, based on a rethink-reuse-reduce enquiry approach, enabled connectedness between two noteworthy animal systems in the CL: dairy cows and meat sheep schemes. Certainly, the use of dairy cow feed-leftovers aligned with the short-term supplementation with WS promoted enhanced testicular function, augmented seminal volume, and an increased sexual behavior in Black Belly rams in northern Mexico. Finally, while our research outcomes should enhance not only the resilience and sustainability of sheep production and the well-being of sheep-producers and their families, it may also embrace clinical translational applications.
Collapse
Affiliation(s)
- Andrés J. Rodriguez-Sánchez
- Programa de Posgrado en Ciencias en Producción Agropecuaria, Universidad Autónoma Agraria Antonio Narro Unidad Laguna, Torreon 27054, Mexico
| | - Cesar A. Meza-Herrera
- Unidad Regional Universitaria de Zonas Áridas, Universidad Autónoma Chapingo, Mapimí 35230, Mexico
| | - Angeles De Santiago-Miramontes
- Programa de Posgrado en Ciencias en Producción Agropecuaria, Universidad Autónoma Agraria Antonio Narro Unidad Laguna, Torreon 27054, Mexico
| | - Cayetano Navarrete-Molina
- Departmento de Química Area Tecnología Ambiental, Universidad Tecnológica de Rodeo, Rodeo 35760, Mexico; (C.N.-M.)
| | - Francisco G. Veliz-Deras
- Programa de Posgrado en Ciencias en Producción Agropecuaria, Universidad Autónoma Agraria Antonio Narro Unidad Laguna, Torreon 27054, Mexico
| | - Julieta Z. Ordoñez-Morales
- Programa de Posgrado en Ciencias en Producción Agropecuaria, Universidad Autónoma Agraria Antonio Narro Unidad Laguna, Torreon 27054, Mexico
| | - Jessica M. Flores-Salas
- Programa de Posgrado en Ciencias en Producción Agropecuaria, Universidad Autónoma Agraria Antonio Narro Unidad Laguna, Torreon 27054, Mexico
| | - Ruben I. Marin-Tinoco
- Departmento de Química Area Tecnología Ambiental, Universidad Tecnológica de Rodeo, Rodeo 35760, Mexico; (C.N.-M.)
- Hospital Rural no. 162 Instituto Mexicano del Seguro Social, Rodeo 35760, Mexico
| |
Collapse
|
15
|
Liu Y, Du M, Zhang L, Wang N, He Q, Cao J, Zhao B, Li X, Li B, Bou G, Zhao Y, Dugarjaviin M. Comparative Analysis of mRNA and lncRNA Expression Profiles in Testicular Tissue of Sexually Immature and Sexually Mature Mongolian Horses. Animals (Basel) 2024; 14:1717. [PMID: 38929336 PMCID: PMC11200857 DOI: 10.3390/ani14121717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Testicular development and spermatogenesis are tightly regulated by both coding and non-coding genes, with mRNA and lncRNA playing crucial roles in post-transcriptional gene expression regulation. However, there are significant differences in regulatory mechanisms before and after sexual maturity. Nevertheless, the mRNAs and lncRNAs in the testes of Mongolian horses have not been systematically identified. In this study, we first identified the testicular tissues of sexually immature and sexually mature Mongolian horses at the tissue and protein levels, and comprehensively analyzed the expression profiles of mRNA and lncRNA in the testes of 1-year-old (12 months, n = 3) and 10-year-old (n = 3) Mongolian horses using RNA sequencing technology. Through gene expression analysis, we identified 16,582 mRNAs and 2128 unknown lncRNAs that are commonly expressed in both sexually immature and sexually mature Mongolian horses. Meanwhile, 9217 mRNAs (p < 0.05) and 2191 unknown lncRNAs (p < 0.05) were identified as differentially expressed between the two stages, which were further validated by real-time fluorescent quantitative PCR and analyzed using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). The analysis results showed that genes in the sexually immature stage were mainly enriched in terms related to cellular infrastructure, while genes in the sexually mature stage were enriched in terms associated with hormones, metabolism, and spermatogenesis. In summary, the findings of this study provide valuable resources for a deeper understanding of the molecular mechanisms underlying testicular development and spermatogenesis in Mongolian horses and offer new perspectives for future related research.
Collapse
Affiliation(s)
- Yuanyi Liu
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ming Du
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lei Zhang
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Na Wang
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Qianqian He
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jialong Cao
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Bilig Zhao
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xinyu Li
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Bei Li
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Gerelchimeg Bou
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yiping Zhao
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Manglai Dugarjaviin
- Key Laboratory of Equus Germplasm Innovation, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (Y.L.); (L.Z.); (N.W.); (Q.H.); (J.C.); (B.Z.); (X.L.); (B.L.); (G.B.); (Y.Z.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
16
|
Erthal-Michelato RP, Quadreli DH, Zaninelli TH, Verri WA, Fernandes GSA. Lower malathion concentrations reduce testosterone biosynthesis by Leydig TM3 cells in vitro by altering cellular redox profile and inducing oxidative damage. Reprod Toxicol 2024; 126:108595. [PMID: 38641014 DOI: 10.1016/j.reprotox.2024.108595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Malathion is an organophosphate pesticide used in agriculture and control of the Aedes aegypti mosquito. As previous reports have indicated the potential of malathion to compromise testosterone production in in vivo models, the objective of this study was to elucidate the mechanisms underlying the impairment of Leydig cell function, considering its critical role in male reproductive function. To this end, murine Leydig TM3 cells were exposed to concentrations of 1, 10, 100 or 1000 μM malathion for 24 h for evaluation of the compound on cell viability. Subsequently, concentrations of 1, 10, and 100 μM malathion were employed for a 24-h period to assess testosterone biosynthesis, levels of cytokines IL-1β, IL-6, IL-10, and TNF-α, as well as the redox profile. Malathion exerted a concentration-dependent impact on cell viability. Notably, the lower concentrations of malathion (1 and 10 μM) were found to impair testosterone biosynthesis in TM3 cells. While there were changes in IL-1 and TNF-α levels at specific concentrations, no direct correlation with altered hormone production was established. Our investigation revealed that varied malathion concentrations induced oxidative stress by increase in superoxide anion and a compensatory rise in antioxidants. In conclusion, the observed changes in the oxidative profile of TM3 cells were linked to functional impairment, evidenced by reduced testosterone biosynthesis at lower malathion concentrations.
Collapse
Affiliation(s)
- Rafaela Pires Erthal-Michelato
- Department of General Biology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil; Department of General Pathology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil.
| | - Débora Hipólito Quadreli
- Department of General Biology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil
| | - Tiago Henrique Zaninelli
- Department of General Pathology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil
| | - Waldiceu Aparecido Verri
- Department of General Pathology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil
| | - Glaura Scantamburlo Alves Fernandes
- Department of General Biology, Biological Sciences Center, State University of Londrina - UEL, Rodovia Celso Garcia Cid, PR 445, Londrina, Paraná 86057-970, Brazil
| |
Collapse
|
17
|
Domes G, Linnig K, von Dawans B. Gonads under stress: A systematic review and meta-analysis on the effects of acute psychosocial stress on gonadal steroids secretion in humans. Psychoneuroendocrinology 2024; 164:107004. [PMID: 38471257 DOI: 10.1016/j.psyneuen.2024.107004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Animal research has shown that the hypothalamus-pituitary-gonadal (HPG) axis is inhibited by (chronic and/or severe) stress, which can lead to impaired fertility and reproductive functioning, presumably caused by the inhibition of gonadal steroid secretion and in interactions with glucocorticoids. However, what has not been clarified is how acute psychosocial stress modulates gonadal steroid secretion in humans. Here we summarize the experimental research on the acute effects of stress on the secretion of gonadal steroids in humans. A systematic literature search revealed 21 studies (with N=881 individuals) measuring testosterone, progesterone or estradiol in response to a standardized acute laboratory stressor in healthy humans. Both our literature review and quantitative meta-analysis suggest that in humans, acute stress stimulates rather than inhibits HPG axis activity, although there is a considerable heterogeneity in the reported methods and results. Increased gonadal steroids in response to acute stress contrasts with many animal studies reporting the opposite pattern, at least regarding severe and/or chronic stressors. We discuss methodological issues and challenges for future research and hope to stimulate experimental studies within this area. A better understanding of these mechanisms is needed, and may have important implications for health and disease, as well as the modulation of various behaviors by acute stressors.
Collapse
Affiliation(s)
- Gregor Domes
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany.
| | - Katrin Linnig
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany
| | - Bernadette von Dawans
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany
| |
Collapse
|
18
|
Zhao Y, Luo X, Hu J, Panga MJ, Appiah C, Du Z, Zhu L, Retyunskiy V, Gao X, Ma B, Zhang Q. Syringin alleviates bisphenol A-induced spermatogenic defects and testicular injury by suppressing oxidative stress and inflammation in male zebrafish. Int Immunopharmacol 2024; 131:111830. [PMID: 38520788 DOI: 10.1016/j.intimp.2024.111830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/21/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024]
Abstract
Syringin (SRG) is a bioactive principle possessing extensive activities including scavenging of free radicals, inhibition of apoptosis, and anti-inflammatory properties. However, its effects on spermatogenic defects and testicular injury as well as the underlying mechanisms are still unclear. This study aims to investigate the protective effect of SRG on testis damage in zebrafish and explore its potential molecular events. Zebrafish testicular injury was induced by exposure to bisphenol A (BPA) (3000 μg/L) for two weeks. Fish were treated with intraperitoneal injection of SRG at different doses (5 and 50 mg/kg bodyweight) for two more weeks under BPA induction. Subsequently, the testis and sperm were collected for morphological, histological, biochemical and gene expression examination. It was found that the administration of SRG resulted in a significant protection from BPA-caused impact on sperm concentration, morphology, motility, fertility rate, testosterone level, spermatogenic dysfunction and resulted in increased apoptotic and reactive oxygen species' levels. Furthermore, testicular transcriptional profiling alterations revealed that the regulation of inflammatory response and oxidative stress were generally enriched in differentially expressed genes (DEGs) after SRG treatment. Additionally, it was identified that SRG prevented BPA-induced zebrafish testis injury through upregulation of fn1a, krt17, fabp10a, serpina1l and ctss2. These results indicate that SRG alleviated spermatogenic defects and testicular injury by suppressing oxidative stress and inflammation in male zebrafish.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| | - Xu Luo
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Jinyuan Hu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Mogellah John Panga
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Clara Appiah
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Zhanxiang Du
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Vladimir Retyunskiy
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xing Gao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| | - Qi Zhang
- School of Food Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
19
|
Zimmer J, Mueller L, Frank-Herrmann P, Rehnitz J, Dietrich JE, Bettendorf M, Strowitzki T, Krivega M. Low androgen signaling rescues genome integrity with innate immune response by reducing fertility in humans. Cell Death Dis 2024; 15:30. [PMID: 38212646 PMCID: PMC10784536 DOI: 10.1038/s41419-023-06397-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024]
Abstract
Development of the gonads under complex androgen regulation is critical for germ cells specification. In this work we addressed the relationship between androgens and genomic integrity determining human fertility. We used different study groups: individuals with Differences of Sex Development (DSD), including Complete Androgen Insensitivity Syndrome (CAIS) due to mutated androgen receptor (AR), and men with idiopathic nonobstructive azoospermia. Both showed genome integrity status influenced by androgen signaling via innate immune response activation in blood and gonads. Whole proteome analysis connected low AR to interleukin-specific gene expression, while compromised genome stability and tumorigenesis were also supported by interferons. AR expression was associated with predominant DNA damage phenotype, that eliminated AR-positive Sertoli cells as the degeneration of gonads increased. Low AR contributed to resistance from the inhibition of DNA repair in primary leukocytes. Downregulation of androgen promoted apoptosis and specific innate immune response with higher susceptibility in cells carrying genomic instability.
Collapse
Affiliation(s)
- J Zimmer
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - L Mueller
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - P Frank-Herrmann
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - J Rehnitz
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - J E Dietrich
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - M Bettendorf
- Division of Pediatric Endocrinology, Children's Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - T Strowitzki
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - M Krivega
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
Rizzuti A, Alvarenga C, Stocker G, Fraga L, Santos HO. Early Pharmacologic Approaches to Avert Anabolic Steroid-induced Male Infertility: A Narrative Review. Clin Ther 2023; 45:e234-e241. [PMID: 37806813 DOI: 10.1016/j.clinthera.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE To review the impact of testosterone and other androgenic-anabolic steroids (AASs) on male fertility, exploring potential drugs that can be used to preserve or restore male fertility upon AAS use or prior contact. METHODS A review was performed to provide a unifying clinical link between drugs used to preserve or restore male fertility (ie, clomiphene citrate, human chorionic gonadotropin, selective estrogen receptor modulators, recombinant luteinizing and follicle-stimulating hormones, and human menopausal gonadotrophin) in the context of AAS-induced infertility and related aspects. FINDINGS Human chorionic gonadotropin (125-500 IU every other day), clomiphene citrate (12.5-50 mg/d), recombinant luteinizing hormone (125-500 IU every other day), recombinant follicle-stimulating hormone (75-150 IU 1-3×/wk), and human menopausal gonadotrophin (75-150 IU 1-3×/wk) are promising early pharmacologic approaches to avert AAS-induced male infertility. Additionally, a full partner assessment is crucial to the success of a couple planning to have children. The partner's age and gynecopathies must be considered. Egg or sperm cryopreservation can also be alternatives for future fertility. Reinforcing AAS cessation is imperative to achieving better success in misusers. IMPLICATIONS The exponential increase in AAS misuse raises concerns about the impact on male fertility. This review suggests that gonadotropin analogs and selective androgen receptor modulators (clomiphene citrate) are viable approaches to early preserve or restore fertility in men on AAS use or with previous contact. However, proper standardization of doses and combinations is required and hence physicians should also be aware of patients' and partners' fertility.
Collapse
Affiliation(s)
- André Rizzuti
- School of Medicine, Estácio de Sá University (UNESA), Rio de Janeiro, RJ, Brazil
| | - Conrado Alvarenga
- School of Medicine, Department of Urology, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Gustavo Stocker
- School of Medicine, University Center Assis Gurgacz Foundation (FAG), Cascavel, PR, Brazil
| | - Lucas Fraga
- School of medicine, Santa Casa da misericordia de Vitorica (EMESCAM), Vitória, ES, Brazil
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil.
| |
Collapse
|
21
|
Methorst C, Perrin J, Faix A, Huyghe E. [Male infertility, environment and lifestyle]. Prog Urol 2023; 33:613-623. [PMID: 38012907 DOI: 10.1016/j.purol.2023.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Since the 1970s, there has been a quantitative and qualitative decline in sperm parameters. The main hypothesis to explain such a rapid evolution is the involvement of environmental and behavioral phenomena. METHODS A bibliographic search limited to English and French literature in men published before 7/2023 was carried out on the links between fertility and pollution, xenobiotics, tobacco, narcotics, cannabis, alcohol, weight, sport, sedentary lifestyle, sleep and anabolics. RESULTS Profound changes in lifestyle have occurred over the past 50 years: reduced sleep time, sedentary lifestyle, dietary changes, tobacco consumption, use of narcotics and anabolics. These changes have a proven impact on spermogram parameters, and should be corrected in an effort to optimize reproductive health. Other environmental parameters: pollution, exposure to heavy metals, exposure to xenobiotics, phthalates and pesticides… will be more difficult to exclude from patients' daily lives, but deserve to be taken more into account. CONCLUSION This review should help the urologist to assess and counsel patients in order to improve their reproductive health. These factors should be routinely investigated in infertile men.
Collapse
Affiliation(s)
- C Methorst
- Service de médecine de la reproduction, hôpital des 4 villes, Saint-Cloud, France
| | - J Perrin
- Biologie et médecine de la reproduction et du développement, CHU de Marseille, UMR 7263 IMBE, Marseille, France
| | - A Faix
- Clinique Saint-Roch, 560, avenue du Colonel-Pavelet-dit-Villars, 34000 Montpellier, France
| | - E Huyghe
- Département d'urologie, hôpital de Rangueil, CHU de Toulouse, Toulouse, France; Service de médecine de la reproduction, hôpital Paule-de-Viguier, CHU de Toulouse, Toulouse, France; UMR DEFE, Inserm 1203, université de Toulouse, université de Montpellier, Toulouse, France.
| |
Collapse
|
22
|
Özkan Karasu Y, Orbak R, Kaşalı K, Berker E, Kantarci A. Porphyromonas gingivalis enhances the senescence-induced increase of 5-alpha reductase in gingival fibroblasts. Clin Oral Investig 2023; 27:5977-5989. [PMID: 37608238 DOI: 10.1007/s00784-023-05211-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVES Aging is characterized by chronic inflammatory activity. Senescent cells increase with chronic inflammation and age-related pathologies, including periodontal disease. As a critical regulator of tissue inflammaging, we hypothesized that 5α reductase (5αR) is associated with periodontal disease and bacteria-induced senescence in gingival fibroblasts. MATERIALS AND METHODS We recruited 36 patients with periodontitis, measured 5αR immunohistochemically before and after periodontal treatment, and compared the expression of 5αR in gingival biopsies from 12 healthy individuals. We then tested the impact of Porphyromonas gingivalis on gingival fibroblasts treated with or without D-galactose-induced cell senescence. We treated primary gingival fibroblasts with D-galactose-supplemented media (0 µM, 50 µM, 100 µM, 1 mM, 10 mM, 50 mM) to induce senescence. The expression of type 1 and type 2 5αR was analyzed with real-time PCR and immunocytochemistry. The levels of IL-6, IL-8, TNF-α, and MCP-1 in fibroblast cultures were evaluated by multiplex immunoassay. RESULTS In gingival biopsies from patients with periodontal disease, the expression of 5αR was significantly higher than in samples from individuals without periodontal disease (p < 0.001). Periodontal treatment significantly reduced the expression of 5αR in gingival tissues (p < 0.001) to levels comparable in healthy individuals. Gingival fibroblasts exposed to D-galactose-supplemented media had a dose-dependent and significant increase in 5αR expression (p < 0.001). P. gingivalis caused statistically higher type 1 and type 2 5αR expression in gingival fibroblast cells. This effect was exacerbated by the lower doses of D-galactose (p = 0.037). Cells infected with P. gingivalis produced significantly higher levels of IL-6, IL-8, TNF-α, and MCP-1 (p < 0.05) regardless of the D-galactose exposure. CONCLUSION The results suggested that 5αR plays a role in periodontal disease and mediates the senescence-induced response to P. gingivalis in gingival fibroblasts. CLINICAL RELEVANCE Periodontal diseases and aging can increase the production of 5-alpha reductase in the gingival tissue.
Collapse
Affiliation(s)
- Yerda Özkan Karasu
- The Forsyth Institute, Cambridge, MA, USA
- Faculty of Dentistry, Department of Periodontology, Ataturk University, Erzurum, Turkey
| | - Recep Orbak
- Faculty of Dentistry, Department of Periodontology, Ataturk University, Erzurum, Turkey
| | - Kamber Kaşalı
- Faculty of Medicine, Department of Biostatistics, Ataturk University, Erzurum, Turkey
| | - Ezel Berker
- Faculty of Dentistry, Department of Periodontology, Hacettepe University, Ankara, Turkey
- Faculty of Dentistry, Department of Periodontology, Istanbul Medipol University, Istanbul, Turkey
| | - Alpdogan Kantarci
- The Forsyth Institute, Cambridge, MA, USA.
- School of Dental Medicine, Harvard University, Boston, MA, USA.
| |
Collapse
|
23
|
Eisenberg ML, Esteves SC, Lamb DJ, Hotaling JM, Giwercman A, Hwang K, Cheng YS. Male infertility. Nat Rev Dis Primers 2023; 9:49. [PMID: 37709866 DOI: 10.1038/s41572-023-00459-w] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
Clinical infertility is the inability of a couple to conceive after 12 months of trying. Male factors are estimated to contribute to 30-50% of cases of infertility. Infertility or reduced fertility can result from testicular dysfunction, endocrinopathies, lifestyle factors (such as tobacco and obesity), congenital anatomical factors, gonadotoxic exposures and ageing, among others. The evaluation of male infertility includes detailed history taking, focused physical examination and selective laboratory testing, including semen analysis. Treatments include lifestyle optimization, empirical or targeted medical therapy as well as surgical therapies that lead to measurable improvement in fertility. Although male infertility is recognized as a disease with effects on quality of life for both members of the infertile couple, fewer data exist on specific quantification and impact compared with other health-related conditions.
Collapse
Affiliation(s)
- Michael L Eisenberg
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Obstetrics & Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Sandro C Esteves
- ANDROFERT Andrology and Human Reproduction Clinic, Campinas, Brazil
- Division of Urology, Department of Surgery, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Dolores J Lamb
- Center for Reproductive Genomics, Weill Cornell Medical College, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - James M Hotaling
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kathleen Hwang
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yu-Sheng Cheng
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
24
|
Molina EM, Kavazis AN, Mendonça MT, Akingbemi BT. Effects of chronic dichlorodiphenyldichloroethylene exposure on testosterone secretion and steroidogenic pathway in the male gonad. Biol Reprod 2023; 109:65-72. [PMID: 37104616 DOI: 10.1093/biolre/ioad045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 02/15/2023] [Accepted: 04/26/2023] [Indexed: 04/29/2023] Open
Abstract
Endocrine disrupting chemicals are present in the environment and/or in consumer products. These agents have the capacity to mimic and/or antagonize endogenous hormones and thus perturb the endocrine axis. The male reproductive tract expresses steroid hormone (androgen and estrogen) receptors at high levels and is a major target for endocrine disrupting chemicals. In this study, Long-Evans male rats were exposed to dichlorodiphenyldichloroethylene, a metabolite of dichlorodiphenyltrichloroethane and a chemical present in the environment, in drinking water at 0.1 and 10 μg/L for 4 weeks. At the end of exposure, we measured steroid hormone secretion and analyzed steroidogenic proteins, including 17β-hydroxysteroid dehydrogenase, 3β-hydroxysteroid dehydrogenase, steroidogenic acute regulatory protein, aromatase, and the LH receptor. We also analyzed Leydig cell apoptosis (poly-(ADP-ribose) polymerase) and caspase-3 in the testes. Testicular testosterone (T) and 17β-estradiol (E2) were both affected by exposure to dichlorodiphenyldichloroethylene by displaying altered steroidogenic enzyme expression. Dichlorodiphenyldichloroethylene exposure also increased the expression of enzymes mediating the pathway for programmed cell death, including caspase 3, pro-caspase 3, PARP, and cleaved PARP. Altogether, the present results demonstrate that dichlorodiphenyldichloroethylene directly and/or indirectly can target specific proteins involved in steroid hormone production in the male gonad and suggest that exposure to environmentally relevant dichlorodiphenyldichloroethylene levels has implications for male reproductive development and function.
Collapse
Affiliation(s)
- Erica M Molina
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | | | - Mary T Mendonça
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | - Benson T Akingbemi
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, USA
| |
Collapse
|
25
|
Bui-Le TN, Hoang-Tan Q, Hoang-Viet H, Truong-Thi BP, Nguyen-Thanh T. Protective Effect of Curculigo orchioides Gaertn. Extract on Heat Stress-Induced Spermatogenesis Complications in Murine Model. Curr Issues Mol Biol 2023; 45:3255-3267. [PMID: 37185736 PMCID: PMC10136419 DOI: 10.3390/cimb45040212] [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: 02/27/2023] [Revised: 03/27/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Curculigo orchioides Gaertn. is a precious herb used in traditional medicine systems in Asian countries for various health benefits. This study investigated the potential protective effects of C. orchioides extract on reproductive health under heat stress conditions in male mice. Forty-eight mice were divided into eight groups, control condition (C group), C. orchioides extract at the dosages of 100, 200, and 400 mg/kg/day (C100, C200, C400 group), 40 °C heat exposure (H group), and combined 40 °C heat exposure and C. orchioides extract at the dosages of 100, 200, and 400 mg/kg/day (HC100, HC200, HC400 group). The result shows that the mice that received only C. orchioides extract without heat stress do not have a significant change in histological structure and testosterone level. The histological analysis of testicular tissue showed that heat stress conditions reduced reproductive function and inhibited the spermatogenesis of male mice. The C. orchioides rhizome extract treatment attenuated the heat stress-induced spermatogenesis complications in the murine model. Mice in the heat-stress group treated with C. orchioides extract had increased spermatogenic cells and spermatozoa compared with mice exposed to heat without C. orchioides treatment. Moreover, the aqueous extract of C. orchioides rhizome enhanced the serum total testosterone levels in heat-exposed mice. In conclusion, the study findings validate that C. orchioides is effective against heat stress-induced spermatogenesis complications in the murine model.
Collapse
Affiliation(s)
- Thanh-Nhan Bui-Le
- Faculty of Basic Science, University of Medicine and Pharmacy, Hue University, Hue 49000, Vietnam
- Faculty of Biology, University of Sciences, Hue University, Hue 49000, Vietnam
| | - Quang Hoang-Tan
- Institute of Biotechnology, Hue University, Hue 49000, Vietnam
| | - Huong Hoang-Viet
- Thua Thien Hue Department of Science and Technology, Hue 49000, Vietnam
| | | | - Tung Nguyen-Thanh
- Faculty of Basic Science, University of Medicine and Pharmacy, Hue University, Hue 49000, Vietnam
- Institute of Biomedicine, University of Medicine and Pharmacy, Hue University, Hue 49000, Vietnam
| |
Collapse
|
26
|
Lima LAR, Torres SM, Macêdo SRB, Tenorio FDCAM, Tenorio BM, Amaro da Silva Junior V. Olanzapine treatment of lactating females causes testicular atrophy in prepuberal rat offspring. Biotech Histochem 2023; 98:179-186. [PMID: 36475412 DOI: 10.1080/10520295.2022.2150314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The antipsychotic drug, olanzapine, is prescribed for postpartum psychosis. Possible adverse effects on fertility of offspring are unclear. We investigated the effects of administering olanzapine via lactation on testicular development and endocrine function of prepuberal male rats. Olanzapine was administered to mothers at 2.5, 5 or 10 mg/kg. We found in male offspring increased body weight, decreased gonadosomatic index, testicular weight and epididymal weight. The volume of seminiferous tubules, seminiferous epithelium, Leydig cells, intertubule tissue and lymphatic space was reduced in rat pups exposed to olanzapine. Tubule diameter and length, seminiferous epithelium height, Leydig cell size and nuclear diameter also were reduced. Testosterone levels were reduced in the groups exposed to olanzapine, while prolactin levels were increased. We observed histopathology in testes of animals whose mothers had been treated with 2.5 mg/kg olanzapine; more severe pathology was observed in offspring whose mothers were administered higher doses. Administration of olanzapine to mothers during lactation produced testicular and endocrine pathology in prepuberal rats in a dose-dependent manner.
Collapse
Affiliation(s)
| | - Sandra Maria Torres
- Department of Veterinary Medicine, Federal Rural University of Pernambuco, Recife, Brazil
| | | | | | - Bruno Mendes Tenorio
- Department of Histology and Embryology, Federal University of Pernambuco, Recife, Brazil
| | | |
Collapse
|
27
|
Duan H, Ge W, Wu J, Lv J, Li Z, Dong W, Du X, Zhang L, Zhang Y, Hu J, Zhao X. Melatonin regulates dihydrotestosterone formation via its membrane receptor in the epididymal epithelial cells of sheep. Theriogenology 2023; 198:273-281. [PMID: 36623430 DOI: 10.1016/j.theriogenology.2022.12.040] [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: 02/13/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Both melatonin and androgen, which affect sperm fertility, are the important factors in epididymis of male animal. In the present study, we confirmed that melatonin regulates the formation of dihydrotestosterone (DHT) in sheep epididymides. Here, we investigated the localization and the expression levels of melatonin keys synthases AANAT and HIOMT, membrane receptors MT1 and MT2, and nuclear receptor RORα in sheep epididymides and testes. We also cultured epididymal epithelial cells and treated them with different concentrations of melatonin (10-11-10-7 M) and luzindole (10-5 M) and 4P-PDOT (10-5 M) to investigate whether melatonin is involved in the regulation of DHT formation and whether these effects are mediated through its receptor pathways. The results showed that AANAT, HIOMT, MT1, MT2, and RORα were differentially expressed between sheep epididymides and testes. In addition, melatonin is involved in mediating the formation of DHT in epididymal epithelial cells, and its influence on DHT is at least partially regulated by the melatonin receptor pathway. Our findings showed that melatonin regulates the functions of the testes and epididymides through an autocrine mechanism and regulates the formation of androgen in sheep epididymides via the receptor pathway. These results provide a basis for further exploring the regulatory mechanisms of melatonin in animal reproduction.
Collapse
Affiliation(s)
- Hongwei Duan
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Wenbo Ge
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China; Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Animal Science and Veterinary Pharmaceutics, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, PR China
| | - Jianxin Wu
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Jianshu Lv
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Zongshuai Li
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Weitao Dong
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Xianghong Du
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Lihong Zhang
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Yong Zhang
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China
| | - Junjie Hu
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Xingxu Zhao
- Key Lab of Animal Generational Physiology and Reproductive Regulation of Gansu Province, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| |
Collapse
|
28
|
Esposito M, Salerno M, Calvano G, Agliozzo R, Ficarra V, Sessa F, Favilla V, Cimino S, Pomara C. Impact of anabolic androgenic steroids on male sexual and reproductive function: a systematic review. Panminerva Med 2023; 65:43-50. [PMID: 35146992 DOI: 10.23736/s0031-0808.22.04677-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Anabolic-androgenic steroids (AASs) are a complex cluster of synthetic derivatives of testosterone. AAS abuse is considered a major public health issue since it has increased among young/adolescent males. The use of steroids has a prevalence rate of 14% in young athletes and 30-75% in professional athletes or bodybuilders. AASs simulate the testosterone mechanism, binding the intracellular androgen receptor, and dysregulating the normal hypothalamic-pituitary-gonadal axis in the same way as exogenous testosterone. Abuse can produce several side effects on organs, such as the genital system. The physio-pathological mechanisms that cause AAS abuse-related, genital system disorders in humans are still not completely known. EVIDENCE ACQUISITION This study focuses on the effect of AASs on the male reproductive organs in humans and animals. EVIDENCE SYNTHESIS A systematic review was performed using SCOPUS, PubMed, Google Scholar, and Web of Sciences database up to 31 December 2021 using the keywords: "anabolic-androgenic steroids," "erectile dysfunction," "spermatogenesis" and "infertility;" (anabolic agents) "erectile dysfunction," "spermatogenesis" and "infertility." The review of the literature identified 66 articles published until 2021. Sixty-two articles were included. The use of AASs induces testicular atrophy and azoospermia known as "anabolic steroid-induced hypogonadism." Anabolic steroid induced infertility is characterized by oligo or azoospermia and abnormalities in sperm motility and morphology. Although sperm quality recovers in most cases within 4 months of stopping anabolic steroid abuse, the negative consequences on spermatogenesis can take up to 3 years to disappear. Human studies reported a positive correlation between AAS abuse in athletes and an increase in morphologically abnormal spermatozoa. Animal studies showed the destruction of Leydig cells and testicular atrophy in animals treated with cycles of AASs. CONCLUSIONS The present review of the literature highlights how little is known about the action of AASs on the male genital system. However, although their use is prohibited in many countries, the black market for these substances is still very frequent. The scientific landscape still has a lot to invest in the research of AAS on the male genital system to make young people even more aware of the negative aspects of these substances, contributing to the reduction of these products in an inappropriate way.
Collapse
Affiliation(s)
- Massimiliano Esposito
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Monica Salerno
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Gianluca Calvano
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Roberta Agliozzo
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Vincenzo Ficarra
- Section of Urology, Department of Human and Pediatric Pathology Gaetano Barresi, University of Messina, Messina, Italy
| | - Francesco Sessa
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy
| | - Vincenzo Favilla
- Section of Urology, Department of Human and Pediatric Pathology Gaetano Barresi, University of Messina, Messina, Italy
| | - Sebastiano Cimino
- Section of Urology, Department of Surgery, University of Catania, Catania, Italy
| | - Cristoforo Pomara
- Unit of Legal Medicine, Department of Medical, Surgical and Advanced Technologies, "G.F. Ingrassia, " University of Catania, Catania, Italy -
| |
Collapse
|
29
|
Boulicault M, Perret M, Galka J, Borsa A, Gompers A, Reiches M, Richardson S. The future of sperm: a biovariability framework for understanding global sperm count trends. HUM FERTIL 2022; 25:888-902. [PMID: 33969777 DOI: 10.1080/14647273.2021.1917778] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The past 50 years have seen heated debate in the reproductive sciences about global trends in human sperm count. In 2017, Levine and colleagues published the largest and most methodologically rigorous meta-regression analysis to date and reported that average total sperm concentration among men from 'Western' countries has decreased by 59.3% since 1973, with no sign of halting. These results reverberated in the scientific community and in public discussions about men and masculinity in the modern world, in part because of scientists' public-facing claims about the societal implications of the decline of male fertility. We find that existing research follows a set of implicit and explicit assumptions about how to measure and interpret sperm counts, which collectively form what we term the Sperm Count Decline hypothesis (SCD). Using the study by Levine and colleagues, we identify weaknesses and inconsistencies in the SCD, and propose an alternative framework to guide research on sperm count trends: the Sperm Count Biovariability hypothesis (SCB). SCB asserts that sperm count varies within a wide range, much of which can be considered non-pathological and species-typical. Knowledge about the relationship between individual and population sperm count and life-historical and ecological factors is critical to interpreting trends in average sperm counts and their relationships to health and fertility.
Collapse
Affiliation(s)
- Marion Boulicault
- Department of Philosophy and Linguistics, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Philosophy, University of Adelaide, Adelaide, Australia
| | - Meg Perret
- Department of the History of Science, Harvard University, Cambridge, MA, USA
| | - Jonathan Galka
- Department of the History of Science, Harvard University, Cambridge, MA, USA
| | - Alex Borsa
- Department of Sociomedical Sciences, Columbia University, New York, NY, USA
| | - Annika Gompers
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Meredith Reiches
- Department of Anthropology, University of Massachusetts, Boston, MA, USA
| | - Sarah Richardson
- Department of the History of Science, Harvard University, Cambridge, MA, USA.,Committee on Degrees in Studies of Women, Gender, and Sexuality, Harvard University, Cambridge, MA, USA
| |
Collapse
|
30
|
Azhar NA, Paul BT, Jesse FFA, Chung ELT, Kamarulrizal MI, Mohd Lila MA. Seminal and histopathological alterations in bucks challenged with Mannheimia haemolytica serotype a2 and its LPS endotoxin. Trop Anim Health Prod 2022; 54:265. [PMID: 35962250 DOI: 10.1007/s11250-022-03262-z] [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: 01/30/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Pneumonic mannheimiosis is a widespread respiratory bacterial disease of small ruminants caused by Mannheimia haemolytica serotype A2. The disease is known to affect the respiratory organs of infected animals, but its effect on other vital and reproductive organs has not been fully explored. Previous studies have demonstrated increased serum pro-inflammatory cytokine concentration post-challenge with M. haemolytica A2 and its LPS, indicating systemic inflammation in the host. This study determined the potential tissue changes and alterations of sperm parameters due to infection of M. haemolytica A2 and its LPS endotoxin. In this study, twelve experimental bucks were randomly assigned to three groups of four bucks each: group 1 (control group) were intranasally inoculated with 2 mL of PBS pH 7.0, group 2 received 2 mL of 1.2 × 109 CFU/mL M. haemolytica A2 intranasally, and group 3 received 2 mL of LPS extracted from 1.2 × 109 CFU/mL of M. haemolytica A2 intravenously. Semen samples were collected at pre-determined intervals using an electro-ejaculator and analysed immediately after collection. All experimental bucks were slaughtered via exsanguination on day 60 to collect their vital and reproductive organs at necropsy, and the samples were processed and analysed for histopathological changes. The current study has revealed that bucks challenged with M. haemolytica A2 and its LPS exhibited alterations in semen parameters such as motility, wave pattern, viability, and morphological abnormalities. Mild to moderate histopathological changes of the lung, liver, testis, epididymis, vas deferens, prostate, and lymph nodes were also observed in both challenged groups. Therefore, this study revealed the potential harmful effects of respiratory mannheimiosis on the reproductive organs of the infected bucks and sheds light on the expanse of systemic effects of this disease.
Collapse
Affiliation(s)
- Nur Amira Azhar
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia
| | - Bura Thlama Paul
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.,Veterinary Teaching Hospital, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri, 600230, Borno State, Nigeria
| | - Faez Firdaus Abdullah Jesse
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia. .,Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Eric Lim Teik Chung
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia.,Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia
| | - Mat Isa Kamarulrizal
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Azmi Mohd Lila
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia
| |
Collapse
|
31
|
Calvert JK, Fendereski K, Ghaed M, Bearelly P, Patel DP, Hotaling JM. The male infertility evaluation still matters in the era of high efficacy assisted reproductive technology. Fertil Steril 2022; 118:34-46. [PMID: 35725120 DOI: 10.1016/j.fertnstert.2022.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 05/04/2022] [Indexed: 11/04/2022]
Abstract
Today's reproductive endocrinology and infertility providers have many tools at their disposal when it comes to achieving pregnancy. In the setting of highly efficacious assisted reproductive technology, it is natural to assume that male factor infertility can be overcome by acquiring sperm and then bypassing the male evaluation. In this review, we go through guideline statements and a stepwise male factor infertility evaluation to propose that a thorough male evaluation remains important to optimize pregnancy and live birth. The foundation of this parallel evaluation is referral to a reproductive urologist for the optimization of the male partner, for advanced diagnostics and interventions, and for the detection of other underlying male pathology. We also discuss what future developments might have an impact on the workup of the infertile male.
Collapse
Affiliation(s)
- Joshua K Calvert
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah
| | - Kiarad Fendereski
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah
| | - Mohammadali Ghaed
- Urology Department, Rasool Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Priyanka Bearelly
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah
| | - Darshan P Patel
- Department of Urology, University of California San Diego Health, San Diego, California
| | - James M Hotaling
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah.
| |
Collapse
|
32
|
Chauhan S, Srivastava MK, Pathak AK. Effect of standardized root extract of ashwagandha ( Withania somnifera) on well-being and sexual performance in adult males: A randomized controlled trial. Health Sci Rep 2022; 5:e741. [PMID: 35873404 PMCID: PMC9297375 DOI: 10.1002/hsr2.741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/27/2022] [Accepted: 07/07/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Aim In Ayurveda, ashwagandha is a popular plant for promoting youthful energy, longevity, and overall well-being. It is also an excellent aphrodisiac herb that aids in the improvement and maintenance of normal sexual health. The present study aims to evaluate the effect of ashwagandha root extract on improving sexual health in adult males. Methods In this 8-week randomized, double-blind, placebo-controlled study, we investigated the aphrodisiac property of an ashwagandha root extract in adult males. Fifty participants with lower sexual desire were randomly allocated to take 300 mg of ashwagandha root extract or placebo capsules twice daily. Outcomes were measured using the derogatis interview for sexual functioning-male (DISF-M) questionnaire, serum testosterone, serum prolactin, and short-form survey-36 quality of life questionnaire before and after the intervention. Results Compared to placebo, ashwagandha root extract supplementation was associated with a statistically significant increase in the total DISF-M scores (mean difference -9.8; 95% confidence interval, -10.73 to -8.87; p < 0.0001; t-test). It was also associated with a statistically significant increase in serum testosterone levels (-66.52; -80.70 to -52.34; p < 0.0001; t-test). However, the prolactin level did not change after intervention in both the ashwagandha and placebo groups (-1.06; -2.78 to 0.66; p > 0.05). Conclusion These findings suggest that ashwagandha demonstrated a significant subjective perception of sexual well-being and assisted in increasing serum testosterone levels in the participants.
Collapse
Affiliation(s)
- Sanjaya Chauhan
- Department of PharmacologyNarayana Hrudayalaya Allied Health SciencesBangaloreKarnatakaIndia
| | - Manoj K. Srivastava
- Department of PsychiatryOm Surgical Centre and Maternity CentreVaranasiUttar PradeshIndia
| | - Anklesh K. Pathak
- Department of Clinical ResearchOm Research CentreLucknowUttar PradeshIndia
| |
Collapse
|
33
|
Desai A, Yassin M, Cayetano A, Tharakan T, Jayasena CN, Minhas S. Understanding and managing the suppression of spermatogenesis caused by testosterone replacement therapy (TRT) and anabolic–androgenic steroids (AAS). Ther Adv Urol 2022; 14:17562872221105017. [PMID: 35783920 PMCID: PMC9243576 DOI: 10.1177/17562872221105017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Use of testosterone replacement therapy (TRT) and anabolic–androgenic steroids
(AAS) has increased over the last 20 years, coinciding with an increase in men
presenting with infertility and hypogonadism. Both agents have a detrimental
effect on spermatogenesis and pose a clinical challenge in the setting of
hypogonadism and infertility. Adding to this challenge is the paucity of data
describing recovery of spermatogenesis on stopping such agents. The unwanted
systemic side effects of these agents have driven the development of novel
agents such as selective androgen receptor modulators (SARMs). Data showing
natural recovery of spermatogenesis following cessation of TRT are limited to
observational studies. Largely, these have shown spontaneous recovery of
spermatogenesis after cessation. Contemporary literature suggests the time frame
for this recovery is highly variable and dependent on several factors including
baseline testicular function, duration of drug use and age at cessation. In some
men, drug cessation alone may not achieve spontaneous recovery, necessitating
hormonal stimulation with selective oestrogen receptor modulators
(SERMs)/gonadotropin therapy or even the need for assisted reproductive
techniques. However, there are limited prospective randomized data on the role
of hormonal stimulation in this clinical setting. The use of hormonal
stimulation with agents such as gonadotropins, SERMs, aromatase inhibitors and
assisted reproductive techniques should form part of the counselling process in
this cohort of hypogonadal infertile men. Moreover, counselling men regarding
the detrimental effects of TRT/AAS on fertility is very important, as is the
need for robust randomized studies assessing the long-term effects of novel
agents such as SARMs and the true efficacy of gonadotropins in promoting
recovery of spermatogenesis.
Collapse
Affiliation(s)
- Ankit Desai
- Department of Andrology, Imperial Healthcare NHS Trust, Charing Cross Hospital, London W6 8RF, UK
| | - Musaab Yassin
- Department of Andrology, Imperial Healthcare NHS Trust, London, UK
| | - Axel Cayetano
- Department of Andrology, Imperial Healthcare NHS Trust, London, UK
| | - Tharu Tharakan
- Department of Andrology, Imperial Healthcare NHS Trust, London, UK
| | - Channa N. Jayasena
- Department of Reproductive Endocrinology, Imperial Healthcare NHS Trust, London, UK
| | - Suks Minhas
- Department of Andrology, Imperial Healthcare NHS Trust, London, UK
| |
Collapse
|
34
|
Gołyszny M, Obuchowicz E, Zieliński M. Neuropeptides as regulators of the hypothalamus-pituitary-gonadal (HPG) axis activity and their putative roles in stress-induced fertility disorders. Neuropeptides 2022; 91:102216. [PMID: 34974357 DOI: 10.1016/j.npep.2021.102216] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022]
Abstract
Neuropeptides being regulators of the hypothalamus-pituitary-adrenal (HPA) axis activity, also affect the function of the hypothalamus-pituitary-gonadal (HPG) axis by regulating gonadotrophin-releasing hormone (GnRH) secretion from hypothalamic neurons. Here, we review the available data on how neuropeptides affect HPG axis activity directly or indirectly via their influence on the HPA axis. The putative role of neuropeptides in stress-induced infertility, such as polycystic ovary syndrome, is also described. This review discusses both well-known neuropeptides (i.e., kisspeptin, Kp; oxytocin, OT; arginine-vasopressin, AVP) and more recently discovered peptides (i.e., relaxin-3, RLN-3; nesfatin-1, NEFA; phoenixin, PNX; spexin, SPX). For the first time, we present an up-to-date review of all published data regarding interactions between the aforementioned neuropeptide systems. The reviewed literature suggest new pathophysiological mechanisms leading to fertility disturbances that are induced by stress.
Collapse
Affiliation(s)
- Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| | - Michał Zieliński
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18 Street, 40-752 Katowice, Poland.
| |
Collapse
|
35
|
Sidhom K, Panchendrabose K, Mann U, Patel P. An update on male infertility and intratesticular testosterone-insight into novel serum biomarkers. Int J Impot Res 2022; 34:673-678. [PMID: 34987179 DOI: 10.1038/s41443-021-00507-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/24/2021] [Accepted: 11/19/2021] [Indexed: 11/09/2022]
Abstract
Intratesticular testosterone is vital for spermatogenesis, male fertility, and virility. Currently the only method to assess levels of intratesticular testosterone is to perform testicular biopsy which is invasive and can lead to several complications. Approaches to assess intratesticular testosterone have been understudied but hold promise as future male contraceptive agents and may grant the ability to monitor patients undergoing hormonal changes from therapeutic and diagnostic perspectives. Previous studies have sought to assess the utility of 17-hydroxyprogesterone (17-OHP) and insulin-like factor 3 (INSL3) as accurate surrogate biomarkers of intratesticular testosterone. The aim of this review is thus to highlight the importance of intratesticular testosterone and the consequent advances that have been made to elucidate the potential of biomarkers for intratesticular testosterone in the context of male infertility.
Collapse
Affiliation(s)
- Karim Sidhom
- Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | - Uday Mann
- Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | - Premal Patel
- Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada. .,Section of Urology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
36
|
Bisphenol S exposure induces cytotoxicity in mouse Leydig cells. Food Chem Toxicol 2022; 160:112805. [PMID: 34990787 DOI: 10.1016/j.fct.2021.112805] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/23/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
Bisphenol S (BPS), an increasingly used alternative to bisphenol A, has been linked to testosterone deficiency and male reproductive dysfunction in laboratory animals. This study aimed to examine the cytotoxicity of BPS exposure to Leydig cells and to investigate its possible mechanisms. After treatment with BPS (100, 200 and 400 μM) for 48 h in vitro, TM3 mouse Leydig cells exhibited a dose-dependent decrease in the viability. Furthermore, BPS challenge triggered oxidative stress manifested by compromised activities of superoxide dismutase and catalase with exaggerated formation of reactive oxygen species. Especially, BPS exposure resulted in augmented mitochondrial permeability transition pore opening, dissipated mitochondrial membrane potential and reduced ATP generation, along with an altered energy metabolism. Moreover, BPS stimulation enhanced BAX expression and caspase-3 activity and inhibited BCL-2 expression. In addition, BPS-treated TM3 cells showed an accumulation of autophagic vacuoles, together with increased Beclin1 and P62 expression and elevated LC3B-II/LC3B-I ratio. These results demonstrated that in vitro exposure to BPS exerted cytotoxicity to TM3 Leydig cells through inducing oxidative stress, mitochondrial impairment, autophagic disturbance and apoptosis.
Collapse
|
37
|
OUP accepted manuscript. Toxicol Res (Camb) 2022; 11:426-436. [PMID: 35782652 PMCID: PMC9244228 DOI: 10.1093/toxres/tfac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/28/2022] [Accepted: 04/05/2022] [Indexed: 11/14/2022] Open
Abstract
Objective The central objective of this study was to investigate the cumulative effects restraint stress and sodium arsenite on reproductive health in male rats. Methods Healthy male Wistar rats were allocated into 4 groups (n = 8). Animals in group 1 served as controls and did not subjected to any stress. Rats in groups 2, 3, and 4 were subjected to either restraint stress (5 h/day) or maintained on arsenic (25 ppm) via drinking water or both for 65 days. After completion of the experimental period, all the rats were analyzed for selected reproductive endpoints. Results Restraint stress or sodium arsenite treatment increased serum corticosterone levels, reduced testicular daily sperm count, epididymal sperm viability, motility, membrane integrity, and decreased testicular steroidogenic enzymes such as 3β- and 17β-hydroxysteroid dehydrogenases associated with reduced serum testosterone levels, deteriorated testicular architecture, and reduced activity levels of testicular superoxide dismutase and catalase accompanied by elevated lipid peroxidation levels. In rats subjected to restraint stress and sodium arsenite, a significant decrease in selected sperm qualitative and quantitative parameters, serum testosterone levels were observed as compared with rats subjected to sodium arsenite alone. A significant increase in the levels of lipid peroxidation with a concomitant decrease in the activities of antioxidant enzymes was observed in the testis of rats subjected to both restraint stress and sodium arsenite treatment as compared with sodium arsenite alone intoxicated rats. Surprisingly, serum corticosterone levels were significantly elevated in rats following both stressors as compared with arsenic alone treated rats. Analysis of atomic absorption spectroscopy revealed that the accumulation of arsenic in the testis of arsenic-treated and arsenic plus immobilization stress groups was significant as compared with controls. Conclusions Based on the findings, it can be concluded that deterioration of male reproductive health could be accelerated in arsenic intoxicated rats following restraint stress.
Collapse
|
38
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
39
|
Endocrinopathies and Male Infertility. LIFE (BASEL, SWITZERLAND) 2021; 12:life12010010. [PMID: 35054403 PMCID: PMC8779600 DOI: 10.3390/life12010010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023]
Abstract
Male infertility is approaching a concerning prevalence worldwide, and inflicts various impacts on the affected couple. The hormonal assessment is a vital component of male fertility evaluation as endocrine disorders are markedly reversible causatives of male infertility. Precise hormonal regulations are prerequisites to maintain normal male fertility parameters. The core male reproductive event, spermatogenesis, entails adequate testosterone concentration, which is produced via steroidogenesis in the Leydig cells. Physiological levels of both the gonadotropins are needed to achieve normal testicular functions. The hypothalamus-derived gonadotropin-releasing hormone (GnRH) is considered the supreme inducer of the gonadotropins and thereby the subsequent endocrine reproductive events. This hypothalamic–pituitary–gonadal (HPG) axis may be modulated by the thyroidal or adrenal axis and numerous other reproductive and nonreproductive hormones. Disruption of this fine hormonal balance and their crosstalk leads to a spectrum of endocrinopathies, inducing subfertility or infertility in men. This review article will discuss the most essential endocrinopathies associated with male factor infertility to aid precise understanding of the endocrine disruptions-mediated male infertility to encourage further research to reveal the detailed etiology of male infertility and perhaps to develop more customized therapies for endocrinopathy-induced male infertility.
Collapse
|
40
|
Fatima I, Qureshi IZ. Intraperitoneal kisspeptin-10 administration ameliorates sodium arsenite-induced reproductive toxicity in adult male mice. Andrologia 2021; 54:e14347. [PMID: 34897760 DOI: 10.1111/and.14347] [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: 09/17/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 11/28/2022] Open
Abstract
The current study investigated the protective ameliorative effect of intraperitoneally administered kisspeptin-10 (50 nmol/day) against reproductive toxicity in adult male mice challenged with 35 days of exposure to sodium arsenite in drinking water. Mice were divided into tap water control, sodium arsenite-alone (4 ppm and 10 ppm), kisspeptin-alone (intermittent and continuous) and combined (sodium arsenite +kisspeptin-10 intermittent and continuous) treatment groups. Results revealed protective effect of both intermittent and continuous kisspeptin doses on reproductive organs against sodium arsenite-induced toxicity. This was indicated by an increase (p < 0.001) in the activity of antioxidant enzymes and a decrease (p < 0.001) in the levels of oxidative stress biomarkers. Concomitant significant increase was noticeable in the relative organ weight (p < 0.01), and serum testosterone and seminal fructose (p < 0.001), and a significant improvement in sperm parameters was also observed. A significant downregulation of lactate dehydrogenase concentration demonstrated further the protective effect of kisspeptin against tissue damage. Histologically, both treatment regimens of kisspeptin combined with sodium arsenite exposure prevented massive germ cell loss and tissue damage, a condition prominent in sodium arsenite-alone-treated mice. The study demonstrates for the first time kisspeptin's potential to mitigate the biochemical and histotoxic effects of arsenic on male reproductive system.
Collapse
Affiliation(s)
- Iffat Fatima
- Department of Zoology (Animal Sciences), Quaid-i-Azam University, Islamabad, Pakistan
| | - Irfan Zia Qureshi
- Department of Zoology (Animal Sciences), Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
41
|
Linhares BL, Miranda EP, Cintra AR, Reges R, Torres LO. Use, Misuse and Abuse of Testosterone and Other Androgens. Sex Med Rev 2021; 10:583-595. [PMID: 37051948 DOI: 10.1016/j.sxmr.2021.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION For several decades, testosterone and its synthetic derivatives have been used for anabolic and androgenic purposes. Initially restricted to professional bodybuilders, these substances gradually became more popular with recreational weightlifters. Considering its increasing prevalence, the consumption of anabolic androgenic steroids (AAS) has become a matter of great concern. Although most side effects are mild and reversible, some of them can cause permanent damage or can be potentially life threatening. OBJECTIVES To review and summarize medical literature regarding misuse and abuse of testosterone and other androgens, in order to provide evidence-based information on the main topics related to this subject, such as how to identify and how to deal with these patients, and to elucidate the multiple possible adverse effects secondary to this practice. METHODS Key studies were retrieved from PubMed (1989-2021) with reference searches from relevant articles. Search terms included "hypogonadism", "anabolic androgenic steroids", "androgens", "misuse AND testosterone", "abuse AND testosterone", and "side effects AND testosterone". RESULTS There is a significant lack of information in the peer-reviewed literature describing demographic data, implications for different organ systems and the management of current or former AAS users; however, androgen abuse has been already linked to a wide variety of cardiovascular diseases, metabolic, endocrine, neurological, psychiatric and liver disorders. Despite all this, most physicians still feel uncomfortable and hesitate to discuss the issue with patients. CONCLUSIONS The chronic use of high doses of AAS is associated with adverse effects in several organ systems; however, there are still many gaps in our knowledge about the long-term consequences of this practice and how to deal with these patients. Healthcare professionals have a crucial role in combating this public health problem, recognizing and preventing the spread of androgen abuse. Linhares BL, Miranda EP, Cintra AR, et al. Use, Misuse and Abuse of Testosterone and Other Androgens. Sex Med Rev 2021;XX:XXX-XXX.
Collapse
Affiliation(s)
- Bruno L Linhares
- Division of Urology, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Eduardo P Miranda
- Division of Urology, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Anna R Cintra
- Division of Urology, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Ricardo Reges
- Division of Urology, Universidade Federal do Ceara, Fortaleza, Brazil
| | | |
Collapse
|
42
|
Dong X, Jiang H, Li S, Zhang D. Low Serum Testosterone Concentrations Are Associated With Poor Cognitive Performance in Older Men but Not Women. Front Aging Neurosci 2021; 13:712237. [PMID: 34790110 PMCID: PMC8591394 DOI: 10.3389/fnagi.2021.712237] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Current evidence on the association between serum testosterone and cognitive performance has been inconsistent, especially in older adults. To investigate the associations between serum testosterone and cognitive performance in a nationally representative sample of older men and women. Methods: We used data from the National Health and Nutrition Examination Survey (NHANES) 2011-2014. 1,303 men and 1,349 women aged 60 years or older were included in the study. Serum total testosterone was preformed via isotope dilution liquid chromatography tandem mass spectrometry (ID-LC-MS/MS) method. Free testosterone was calculated by Vermeulen's formula. Cognitive performance was evaluated by the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) test, Animal Fluency test, and Digit Symbol Substitution Test (DSST). Binary logistic regression and restricted cubic spline models were applied to evaluate the association of testosterone and cognitive performance. Results: In men, higher concentrations of total testosterone were associated with better performance on CERAD test (OR = 0.51; 95%CI = 0.27-0.95) and DSST (OR = 0.54; 95%CI = 0.30-0.99) in adjusted group. Similarly, higher concentrations of free testosterone were associated with better performance on CERAD test (OR = 0.32; 95%CI = 0.17-0.61) and DSST (OR = 0.41; 95%CI = 0.17-0.96) in men. These associations were not seen in women. Conclusion: Serum testosterone concentrations were inversely associated with cognitive performance in older men but not women in the United States.
Collapse
Affiliation(s)
- Xue Dong
- Department of Epidemiology and Health Statistics, The School of Public Health, Qingdao University, Qingdao, China
| | - Hong Jiang
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Suyun Li
- Department of Epidemiology and Health Statistics, The School of Public Health, Qingdao University, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Leisegang K, Roychoudhury S, Slama P, Finelli R. The Mechanisms and Management of Age-Related Oxidative Stress in Male Hypogonadism Associated with Non-communicable Chronic Disease. Antioxidants (Basel) 2021; 10:1834. [PMID: 34829704 PMCID: PMC8615233 DOI: 10.3390/antiox10111834] [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: 10/14/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Androgens have diverse functions in muscle physiology, lean body mass, the regulation of adipose tissue, bone density, neurocognitive regulation, and spermatogenesis, the male reproductive and sexual function. Male hypogonadism, characterized by reduced testosterone, is commonly seen in ageing males, and has a complex relationship as a risk factor and a comorbidity in age-related noncommunicable chronic diseases (NCDs), such as obesity, metabolic syndrome, type 2 diabetes, and malignancy. Oxidative stress, as a significant contributor to the ageing process, is a common feature between ageing and NCDs, and the related comorbidities, including hypertension, dyslipidemia, hyperglycemia, hyperinsulinemia, and chronic inflammation. Oxidative stress may also be a mediator of hypogonadism in males. Consequently, the management of oxidative stress may represent a novel therapeutic approach in this context. Therefore, this narrative review aims to discuss the mechanisms of age-related oxidative stress in male hypogonadism associated with NCDs and discusses current and potential approaches for the clinical management of these patients, which may include conventional hormone replacement therapy, nutrition and lifestyle changes, adherence to the optimal body mass index, and dietary antioxidant supplementation and/or phytomedicines.
Collapse
Affiliation(s)
- Kristian Leisegang
- School of Natural Medicine, Faculty of Community and Health Sciences, Bellville, Cape Town 7535, South Africa
| | | | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, 61300 Brno, Czech Republic
| | | |
Collapse
|
44
|
Semaida AI, El-Khashab MA, Saber AA, Hassan AI, Elfouly SA. Effects of Sargassum virgatum extracts on the testicular measurements, genomic DNA and antioxidant enzymes in irradiated rats. Int J Radiat Biol 2021; 98:191-204. [PMID: 34694945 DOI: 10.1080/09553002.2022.1998702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/17/2021] [Accepted: 10/07/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Oxidative stress and reactive oxygen species (ROS) are primarily responsible for the development of male infertility after exposure to γ-irradiation. The present work aimed to assess the ameliorative and therapeutic roles of the aqueous and ethanolic extracts of the edible seaweed Sargassum virgatum (S. virgatum) on spermatogenesis and infertility in γ-irradiated Wistar rats. MATERIALS AND METHODS Induction of infertility was performed by exposing the rats to 137Cs-gamma rays, using a single dose of 3.5 Gy. γ-irradiated rats were given the S. virgatum ethanolic (S. virgatum-EtOH) and aqueous extracts intraperitoneally on a daily base for two consecutive weeks at doses of 100 and 400 mg/kg body weight (b.wt.) for each seaweed extract. Morphometric data of the testes, semen quality indices, antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), and glutathione peroxidase (GPx), and deoxyribonucleic acid (DNA) fragmentation were assessed. The results obtained were taken during two-time intervals of 15 and 60 days from the commencement of the algal treatments. In vitro antioxidant assays and polyphenolic compounds of S. virgatum were characterized. RESULTS Significant negative changes in the semen quality and morphometric data of the testes, as well as remarkable DNA fragmentation, were detected in the irradiated rats compared to the control. The levels of the endogenous antioxidant enzymes (SOD, CAT, GSH, and GPx) were also significantly diminished. Nonetheless, treatments of γ-irradiated rats with the S. virgatum-EtOH and aqueous extracts significantly improved the above-mentioned enzymes, in addition to noteworthy amendments in the dimensions of the testes, the semen quality, as well as the DNA structure. CONCLUSIONS The ameliorative potency of S. virgatum to cure γ-irradiation-induced male infertility, particularly 400 mg/kg ethanolic extract for 60 days, is the result of the consistent therapeutic interventions of its potent antioxidant and anti-apoptotic polyphenols, particularly protocatechuic, p-hydroxybenzoic, rosmarinic, chlorogenic, cinnamic and gentisic acids, as well as the flavonoids catechin, hesperidin, rutin and quercetin. Besides its high-value nutraceutical importance, S. virgatum could be a natural candidate for developing well-accepted radioprotectant products capable of treating γ-irradiation-induced male infertility.
Collapse
Affiliation(s)
- Ahmed I Semaida
- Department of Animal Production (Animal Physiology), Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Mona A El-Khashab
- Department of Animal Production (Animal Physiology), Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Abdullah A Saber
- Botany Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Amal I Hassan
- Department of Radioisotopes, Nuclear Research Centre, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Shady A Elfouly
- Department of Radioisotopes, Nuclear Research Centre, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
45
|
Wang TE, Yeh LY, Kuo-Kuang Lee R, Lu CH, Yang TH, Kuo YW, Joshi R, Tsai PS, Li SH. Secretory mouse quiescin sulfhydryl oxidase 1 aggregates defected human and mouse spermatozoa in vitro and in vivo. iScience 2021; 24:103167. [PMID: 34667943 PMCID: PMC8506963 DOI: 10.1016/j.isci.2021.103167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
A flavin-dependent enzyme quiescin Q6 sulfhydryl oxidase 1 (QSOX1) catalyzes the oxidation of thiol groups into disulfide bonds. QSOX1 is prominently expressed in the seminal plasma. However, its role in male reproduction is elusive. Here, we purified the secreted form of QSOX1, i.e., QSOX1c, from mouse seminal vesicle secretions and revealed for the first time its function involved in sperm physiology. Exogenous addition of QSOX1c time-dependently promoted the in vitro aggregation of thiol-rich, oxidative stressed, and apoptotic mouse and human sperm cells. Also, in vivo aggregated sperm cells collected from mouse uterine and human ejaculates also showed high levels of QSOX1c, intracellular reactive oxygen species, annexin V, and free thiols. In summary, our studies demonstrated that QSOX1c could agglutinate spermatozoa susceptible to free radical attack and apoptosis. This characteristic may provide an opportunity to separate defective sperm cells and improve sperm quality before artificial insemination in humans and animals. QSOX1c is expressed in the seminal vesicle and presented in the seminal plasma QSOX1c agglutinates thiol-rich, oxidatively stressed, and apoptotic sperm QSOX1c aggregates impaired sperm presented in the mouse uterine and human ejaculates QSOX1c-treated semen may improve the sperm quality for artificial insemination
Collapse
Affiliation(s)
- Tse-En Wang
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Ling-Yu Yeh
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Robert Kuo-Kuang Lee
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsung-Hsien Yang
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Yu-Wen Kuo
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Radhika Joshi
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| |
Collapse
|
46
|
Exposure to Atrazine through gestation and lactation period led to impaired sexual maturation and subfertility in F1 male rats with congenital deformities in F2 progeny. Food Chem Toxicol 2021; 157:112586. [PMID: 34600026 DOI: 10.1016/j.fct.2021.112586] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 11/21/2022]
Abstract
Several scientific reports suggest perturbed reproductive and developmental defects associated with environmental exposure to Atrazine (ATR). ATR has been associated with altered endocrine and reproductive functioning in-vivo exposed during the critical window of development. Thus, the present study investigates the effect of ATR exposure on F1-F2 male progeny exposed through gestation and lactation. F0 dams administered with ATR at doses 2, 10, 70, and 100 mg/kg b. wt/day from gestation day 6 to postnatal day 21. The F1 male rats were monitored for sexual maturation and subjected to fertility assessment on PND75. Delayed testicular descent was observed in 10, 70, and 100 mg/kg b. wt/day ATR dose with significantly lower serum testosterone, sperm count, and motility with testicular defects in F1 male. Expression of Androgen receptor (AR), Estrogen receptors (ER α and ER β), StAR, Aromatase, and INSL-3 were upregulated at all doses indicating estrogenic/anti-androgenic activity of ATR. Fertility assessment revealed subfertility in F1 males with high (%) pre- and post-implantation loss at 10, 70, and 100 mg/kg b. wt/day dose as compared to control. Further, F2 fetuses exhibited congenital disabilities viz. decreased weight, crown-rump length, and anogenital distance with several other morphological deformities. To conclude, ATR exerted estrogenic and/or anti-androgenic activity with fetotoxic effects through the male germline.
Collapse
|
47
|
Akinola LK, Uzairu A, Shallangwa GA, Abechi SE. In silico prediction of nuclear receptor binding to polychlorinated dibenzofurans and its implication on endocrine disruption in humans and wildlife. Curr Res Toxicol 2021; 2:357-365. [PMID: 34693345 PMCID: PMC8515090 DOI: 10.1016/j.crtox.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/10/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022] Open
Abstract
Polychlorinated dibenzofurans (PCDFs) are known to cause endocrine disruption in humans and wildlife but the mechanisms underlying this disruption have not been adequately investigated. In this paper, the susceptibility of the endocrine system to disruption by PCDF congeners via nuclear receptor binding was studied using molecular docking simulation. Findings revealed that some PCDF congeners exhibit high probabilities of binding to androgen receptor in its agonistic and antagonistic conformations. In depth molecular docking analysis of the receptor-ligand complexes formed by PCDFs with androgen receptor in its agonistic and antagonistic conformations showed that, these complexes were stabilized by electrostatic, van der Waals, pi-effect and hydrophobic interactions. It was also observed that PCDF molecules mimic the modes of interaction observed in androgen-testosterone and androgen-bicalutamide complexes, utilizing between 65 and 83% of the amino acid residues used by the co-crystallized ligands for binding. This computational study suggests that some PCDF congeners may act as agonists and antagonists of androgen receptor in humans and wildlife via inapproprate binding to the receptor.
Collapse
Affiliation(s)
- Lukman K. Akinola
- Department of Chemistry, Ahmadu Bello University, Zaria, Nigeria
- Department of Chemistry, Bauchi State University, Gadau, Nigeria
| | - Adamu Uzairu
- Department of Chemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | | |
Collapse
|
48
|
Sex Determination and Differentiation in Teleost: Roles of Genetics, Environment, and Brain. BIOLOGY 2021; 10:biology10100973. [PMID: 34681072 PMCID: PMC8533387 DOI: 10.3390/biology10100973] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 01/19/2023]
Abstract
The fish reproductive system is a complex biological system. Nonetheless, reproductive organ development is conserved, which starts with sex determination and then sex differentiation. The sex of a teleost is determined and differentiated from bipotential primordium by genetics, environmental factors, or both. These two processes are species-specific. There are several prominent genes and environmental factors involved during sex determination and differentiation. At the cellular level, most of the sex-determining genes suppress the female pathway. For environmental factors, there are temperature, density, hypoxia, pH, and social interaction. Once the sexual fate is determined, sex differentiation takes over the gonadal developmental process. Environmental factors involve activation and suppression of various male and female pathways depending on the sexual fate. Alongside these factors, the role of the brain during sex determination and differentiation remains elusive. Nonetheless, GnRH III knockout has promoted a male sex-biased population, which shows brain involvement during sex determination. During sex differentiation, LH and FSH might not affect the gonadal differentiation, but are required for regulating sex differentiation. This review discusses the role of prominent genes, environmental factors, and the brain in sex determination and differentiation across a few teleost species.
Collapse
|
49
|
Oxidative Stress, Testicular Inflammatory Pathways, and Male Reproduction. Int J Mol Sci 2021; 22:ijms221810043. [PMID: 34576205 PMCID: PMC8471715 DOI: 10.3390/ijms221810043] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammation is among the core causatives of male infertility. Despite male infertility being a serious global issue, "bits and pieces" of its complex etiopathology still remain missing. During inflammation, levels of proinflammatory mediators in the male reproductive tract are greater than usual. According to epidemiological research, in numerous cases of male infertility, patients suffer from acute or chronic inflammation of the genitourinary tract which typically occurs without symptoms. Inflammatory responses in the male genital system are inextricably linked to oxidative stress (OS). OS is detrimental to male fertility parameters as it causes oxidative damage to reproductive cells and intracellular components. Multifarious male infertility causative factors pave the way for impairing male reproductive functions via the common mechanisms of OS and inflammation, both of which are interlinked pathophysiological processes, and the occurrence of any one of them induces the other. Both processes may be simultaneously found in the pathogenesis of male infertility. Thus, the present article aims to explain the role of inflammation and OS in male infertility in detail, as well as to show the mechanistic pathways that link causative factors of male reproductive tract inflammation, OS induction, and oxidant-sensitive cellular cascades leading to male infertility.
Collapse
|
50
|
Liu S, Tang Y, Chen B, Zhao Y, Aguilar ZP, Tao X, Xu H. Inhibition of testosterone synthesis induced by oral TiO 2 NPs is associated with ROS-MAPK(ERK1/2)-StAR signaling pathway in SD rat. Toxicol Res (Camb) 2021; 10:937-946. [PMID: 34484685 DOI: 10.1093/toxres/tfab077] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) have been widely used in food, medical, and other fields; their reproductive toxicity has been reported in numerous studies. However, the relevant toxicity mechanism still requires further exploration. In this paper, the effect of oral exposure to 500 mg/kg TiO2 NPs (anatase and rutile) in adult male SD rats was studied over 3 and 7 days. Results showed that the total sperm count and testosterone level of 7 days of exposure in serum decreased in the experimental group. Testicular tissue lesions, such as disappearance of Leydig cells, disorder of arrangement of spermatogenic cells in the lumen of convoluted seminiferous tubules, and disorder of arrangement of germ cells, were observed. Meanwhile, the expression of steroidogenic acute regulatory (StAR; the key factors of testosterone synthesis), MAPK (ERK1/2), and phosphorylated ERK1/2 in testes of SD rats after exposure to TiO2 NPs for 7 days decreased, while the malondialdehyde content increased and superoxide dismutase activity decreased in serum. The present study showed that TiO2 NPs could cause reproductive toxicity. Notably, anatase is more toxic than rutile. In addition, exposure to 500 mg/kg TiO2 NPs for 7 days inhibited testosterone synthesis in male rat, which may be related to the reactive oxygen species (ROS)-MAPK (ERK1/2)-StAR signal pathway. Warning that the use of TiO2 NPs should be regulated.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Yizhou Tang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Bolu Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | | | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| |
Collapse
|