1
|
Zhang Y, Shu M, Shan S, Liu H, Zhang Y, Song C, Xu Q, Fan Y, Lu C. Perfluorohexane Sulfonic Acid Disrupts the Immune Microenvironment for Spermatogenesis by Damaging the Structure of the Blood-Testis Barrier in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409383. [PMID: 39823537 PMCID: PMC11904953 DOI: 10.1002/advs.202409383] [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: 08/08/2024] [Revised: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Perfluorohexane sulfonic acid (PFHxS) is extensively used in waterproof coatings and fire-fighting foams, and several studies have found it to be a potential health hazard, but there is still unknown about its effects on spermatogenesis. Our results showed that PFHxS-treated mice have significant reproductive toxicity, including a decrease in sperm count and motility, and the levels of sex hormones (P < 0.05). Concurrently, structural abnormalities are observed in sperm, affecting ≈60-75% of those in the PFHxS-treated group. Additionally, it is found that the structure of the blood-testis barrier (BTB) is damaged after PFHxS treatment, leading to higher expression levels of inflammatory cytokines in the microenvironment for spermatogenesis. Moreover, the expression of proteins associated with mitochondrial biogenesis, including PTEN-induced kinase 1 (PINK1) and NADPH oxidase 4 (NOX4), is dysregulated in the testes after PFHxS treatment. Based on metabolome data, the differential metabolite 3-hydroxybutanoic acid is identified in the PFHxS-treated group, which can regulate the histone Kac levels, especially H3K4ac and H3K9ac. In summary, the results of this study suggest that in the testes of PFHxS-treated mice, inflammatory factors disrupt the mitochondrial function and metabolic profiles and hinder the progress of gene transcription through histone Kac, ultimately causing sperm dysfunction.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
- Department of Epidemiology, School of Public HealthNantong UniversityNantong226001China
| | - Mingxue Shu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Shilin Shan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Huiying Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Yucheng Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Chenyang Song
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Qiaoqiao Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Yun Fan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjing211166China
| |
Collapse
|
2
|
Sugawara T, Sonoda K, Chompusri N, Noguchi K, Okada S, Furuse M, Wakayama T. Claudin-11 regulates immunological barrier formation and spermatogonial proliferation through stem cell factor. Commun Biol 2025; 8:148. [PMID: 39885308 PMCID: PMC11782696 DOI: 10.1038/s42003-025-07592-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025] Open
Abstract
Tight junctions (TJs) between adjacent Sertoli cells are believed to form immunological barriers that protect spermatogenic cells expressing autoantigens from autoimmune responses. However, there is no direct evidence that Sertoli cell TJs (SCTJs) do indeed form immunological barriers. Here, we analyzed male mice lacking claudin-11 (Cldn11), which encodes a SCTJ component, and found autoantibodies against antigens of spermatocytes/spermatids in their sera. Defective spermatogenesis in Cldn11-deficient mice was not restored on a recombination activating gene 2 (Rag2) knockout background lacking mature T and B lymphocytes. This suggests that adaptive immune responses to spermatogenic cells are not a cause of defective spermatogenesis in Cldn11-deficient mice. Further analyses showed that Cldn11 knockout impaired Sertoli cell polarization, localization of stem cell factor (SCF) (a key molecule for maintaining differentiating spermatogonia) to the basal compartment of seminiferous tubules, and also proliferation of differentiating spermatogonia. We propose that CLDN11 creates a microenvironment for SCF-mediated spermatogonial proliferation at the basal compartment via Sertoli cell polarization.
Collapse
Affiliation(s)
- Taichi Sugawara
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Kayoko Sonoda
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Nattapran Chompusri
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuhiro Noguchi
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Aichi, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi, Japan
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiko Wakayama
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Moreira RJ, Oliveira PF, Spadella MA, Ferreira R, Alves MG. Do Lifestyle Interventions Mitigate the Oxidative Damage and Inflammation Induced by Obesity in the Testis? Antioxidants (Basel) 2025; 14:150. [PMID: 40002337 PMCID: PMC11851673 DOI: 10.3390/antiox14020150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity results from a disproportionate accumulation of fat and has become a global health concern. The increase in adipose tissue is responsible for several systemic and testicular changes including hormone levels (leptin, adiponectin, testosterone, estrogen), inflammatory cytokines (increase in TNF-α and IL-6 and decrease in IL-10), and redox state (increase in reactive oxygen species and reduction in antioxidant enzymes). This results in poor sperm quality and compromised fertility in men with obesity. Lifestyle modifications, particularly diet transition to caloric restriction and physical exercise, are reported to reverse these negative effects. Nevertheless, precise mechanisms mediating these benefits, including how they modulate testicular oxidative stress, inflammation, and metabolism, remain to be fully elucidated. The main pathway described by which these lifestyle interventions reverse obesity-induced oxidative damage is the Nrf2-SIRT1 axis, which modulates the overexpression of antioxidant defenses. Of note, some of the detrimental effects of obesity on the testis are inherited by the descendants of individuals with obesity, and while caloric restriction reverses some of these effects, no significant work has been carried out regarding physical exercise. This review discusses the consequences of obesity-induced testicular oxidative stress on adult and pediatric populations, emphasizing the therapeutic potential of lifestyle to mitigate these detrimental effects.
Collapse
Affiliation(s)
- Ruben J. Moreira
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | - Pedro F. Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | | | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (P.F.O.); (R.F.)
| | - Marco G. Alves
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
4
|
Adnane M, Ahmed M, Chapwanya A. Advances in Molecular Biology and Immunology of Spermatozoa and Fertilization in Domestic Animals: Implications for Infertility and Assisted Reproduction. Curr Mol Med 2025; 25:167-186. [PMID: 39572916 DOI: 10.2174/0115665240306965240802075331] [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: 01/27/2024] [Revised: 04/25/2024] [Accepted: 06/13/2024] [Indexed: 04/11/2025]
Abstract
Unlocking the secrets of reproductive success in domestic animals requires a deep understanding of the molecular biology and immunology of spermatozoa, capacitation, fertilization, and conception. This review highlights the complex processes involved in spermatogenesis and sperm capacitation, including changes in membrane properties, signaling pathways, and the crucial acrosome reaction. The interaction with the zona pellucida in species-specific gamete recognition and binding is emphasized. The implications of fertilization defects for infertility and assisted reproduction are discussed, underscoring the challenges faced in breeding programs. The future directions for research in this field involve advancements in molecular techniques, understanding the immune regulation of spermatozoa, investigating environmental factors' impact, and integrating multi-omics approaches to enhance assisted reproduction techniques in domestic animals. This review contributes to our understanding of the intricate mechanisms underlying successful reproduction and provides insights into potential strategies for improving fertility outcomes in domestic animals.
Collapse
Affiliation(s)
- Mounir Adnane
- Department of Biomedicine, Institute of Veterinary Sciences, University Ibn Khaldoun of Tiaret, Tiaret 14000, Algeria
| | - Moussa Ahmed
- Department of Animal Health, Institute of Veterinary Sciences, University Ibn Khaldoun of Tiaret, Tiaret, 14000, Algeria
| | - Aspinas Chapwanya
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre, 00265, Saint Kitts and Nevis
| |
Collapse
|
5
|
Li SY, Kumar S, Gu X, DeFalco T. Testicular immunity. Mol Aspects Med 2024; 100:101323. [PMID: 39591799 PMCID: PMC11624985 DOI: 10.1016/j.mam.2024.101323] [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: 04/02/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
The testis is a unique environment where immune responses are suppressed to allow the development of sperm that possess autoimmunogenic antigens. There are several contributors responsible for testicular immune privilege, including the blood-testis barrier, testicular immune cells, immunomodulation by Sertoli cells, and high levels of steroid hormones. Despite multiple mechanisms in place to regulate the testicular immune environment, pathogens that disrupt testicular immunity can lead to long-term effects such as infertility. If testicular immunity is disturbed, autoimmune reactions can also occur, leading to aberrant immune cell infiltration and subsequent attack of autoimmunogenic germ cells. Here we discuss cellular and molecular factors underlying testicular immunity and how testicular infection or autoimmunity compromise immune privilege. We also describe infections and autoimmune diseases that impact the testis. Further research into testicular immunity will reveal how male fertility is maintained and will help update therapeutic strategies for infertility and other testicular disorders.
Collapse
Affiliation(s)
- Shu-Yun Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sudeep Kumar
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
6
|
Xuan Y, Zhao J, Hong X, Zhang Y, Zhang R, Zhang H, Yan T, Wang Y, Peng Z, Zhang Y, Jiao K, Wang Q, Shen H, Zhang Y, Yan D, Ma X, Wang B. Assessment of male creatinine levels and fecundity in couples planning pregnancy: a national cohort study in China. Hum Reprod 2024; 39:2587-2600. [PMID: 39270672 DOI: 10.1093/humrep/deae206] [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: 04/09/2024] [Revised: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
STUDY QUESTION Is there an association between male creatinine levels and time to pregnancy (TTP) in couples planning pregnancy? SUMMARY ANSWER Low and high male creatinine concentrations were associated with reduced couple fecundity. WHAT IS KNOWN ALREADY Abundant evidence suggests male creatinine dysfunction is associated with infertility in males with kidney diseases. However, the association of preconception creatinine levels with reduced fecundity among general reproductive-aged couples lacks evidence from an in-depth population study. STUDY DESIGN, SIZE, DURATION Based on the population-based cohort study from the National Free Preconception Check-up Projects, 4 023 204 couples were recruited and met the inclusion criteria from 1 January 2015 to 31 December 2017. They were planning pregnancy and were followed up every 3 months until achieving pregnancy as detected by gynaecological ultrasonography or were followed up for 1 year for the analysis of TTP. PARTICIPANTS/MATERIALS, SETTING, METHODS Cox regression models were used to estimate hazard ratios (HRs) and 95% CI for creatinine deciles. Restricted cubic spline regression was adopted for the dose-response relationship of creatinine with HRs. R statistical software was used for data analysis. MAIN RESULTS AND THE ROLE OF CHANCE Of the included participants, 2 756 538 (68.52%) couples successfully conceived. The median male serum creatinine was 81.50 μmol/l. Compared with the reference group (78.00-81.49 μmol/l) including the median creatinine, fecundity in the first (≤64.89 μmol/l), second (64.90-69.99 μmol/l), third (70.00-73.99 μmol/l), and tenth (≥101.00 μmol/l) deciles decreased by 8%, 5%, 2%, and 1%, respectively (Decile 1 Adjusted HR 0.92, 95% CI 0.91-0.92; Decile 2 Adjusted HR 0.95, 95% CI 0.95-0.96; Decile 3 Adjusted HR 0.98, 95% CI 0.97-0.99; Decile 10 Adjusted HR 0.99, 95% CI 0.98-0.99). An inverse-U-shaped association was consistently presented among males such that non-inferiority for fecundity was shown when creatinine was in the 81.66-104.90 μmol/l range (P for non-linearity < 0.001). For males over 40 years old, the risk of fecundity impairment was more obvious and the recommended range of creatinine levels for TTP was reduced and more narrow, compared with that for younger males. LIMITATIONS, REASONS FOR CAUTION Not including the time couples spend preparing for pregnancy before enrolment would lead to an overestimation of fecundity; additionally some couples place pregnancy plans on hold due to special emergencies, which would not have been recognized. Due to the lack of information regarding semen quality, psychological factors, sexual intercourse frequencies, and hazardous environmental factors, we could not adjust for these factors. Some variates were self-reported and dichotomized, which were prone to bias. Direct variables reflecting muscle mass and impaired kidney function were lacking. Thus, extrapolation should be done with caution. WIDER IMPLICATIONS OF THE FINDINGS Male creatinine is associated with couples' fecundity and the relationship varied by age. This study provides a better understanding of the potential implications and significance of different creatinine levels and their association with the clinical significance regarding couples' fecundity. STUDY FUNDING/COMPETING INTEREST(S) This research has received funding from the National Natural Science Foundation of China (Grant No. 81872634), the Basic Research Funds of Central Public Welfare Research Institutes of China (Grant No. 2023GJZ03), the National Key Research and Development Program of China (Grant No. 2016YFC1000307), and the Project of National Research Institute for Family Planning (Grant No. 2018NRIFPJ03), People's Republic of China. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yan Xuan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology and Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Jun Zhao
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Xiang Hong
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology and Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Yue Zhang
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Rong Zhang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology and Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Hongguang Zhang
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Tao Yan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology and Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Yuanyuan Wang
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Zuoqi Peng
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Ya Zhang
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Kailei Jiao
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Qiaomei Wang
- Department of Maternal and Child Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Haiping Shen
- Department of Maternal and Child Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Yiping Zhang
- Department of Maternal and Child Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Donghai Yan
- Department of Maternal and Child Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Bei Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Epidemiology and Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Xu Y, Chen W, Wu X, Zhao K, Liu C, Zhang H. The Role of Cells and Cytokines in Male Infertility Induced by Orchitis. World J Mens Health 2024; 42:681-693. [PMID: 38449458 PMCID: PMC11439807 DOI: 10.5534/wjmh.230270] [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: 09/22/2023] [Revised: 11/05/2023] [Accepted: 11/19/2023] [Indexed: 03/08/2024] Open
Abstract
Recent studies on male infertility reveal a growing worry: more infertile men are dealing with inflammation in the testis. Analyzing testicular biopsies from infertile men highlights a significant presence of inflammation. This connection, supported by clinical and pathological evidence, emphasizes that testicular inflammation hampers sperm production, leading to lasting declines in sperm count and quality. However, the exact reasons behind male infertility due to orchitis, a type of testicular inflammation, are still uncertain. Understanding these fundamental aspects of molecular signals and cellular mechanisms in testicular inflammation is crucial. Our review delves into recent literature with a dual objective: elucidating potential mechanisms involving immune cells, non-immune cells, and cytokines that link orchitis to male infertility, while also paving the way for precise interventions and solutions to address the challenges of male infertility.
Collapse
Affiliation(s)
- Ying Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyi Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Wu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Wei M, Liu H, Wang Y, Sun M, Shang P. Mechanisms of Male Reproductive Sterility Triggered by Dysbiosis of Intestinal Microorganisms. Life (Basel) 2024; 14:694. [PMID: 38929676 PMCID: PMC11204708 DOI: 10.3390/life14060694] [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: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
The intestinal microbiota, comprised of bacteria, archaea, and phages, inhabits the gastrointestinal tract of the organism. Male reproductive sterility is currently a prominent topic in medical research. Increasing research suggests that gut microbiota dysbiosis can result in various reproductive health problems. This article specifically investigates the impact of gut microbiota dysbiosis on male reproductive infertility development. Gut microbiota imbalances can disrupt the immune system and immune cell metabolism, affecting testicular growth and sperm production. This dysfunction can compromise the levels of hormones produced and secreted by the endocrine glands, affecting male reproductive health. Furthermore, imbalance of the gut microbiota can disrupt the gut-brain-reproductive axis, resulting in male reproductive infertility. This article explores how the imbalance of the gut microbiota impacts male reproductive infertility through immune regulation, endocrine regulation, and interactions of the gut-brain-reproductive axis, concluding with recommendations for prevention and treatment.
Collapse
Affiliation(s)
- Mingbang Wei
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (M.W.); (H.L.); (Y.W.); (M.S.)
- The Provincial and Ministerial Co-Founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Linzhi 860000, China
- Key Laboratory for the Genetic Improvement and Reproduction Technology of the Tibetan Swine, Linzhi 860000, China
| | - Huaizhi Liu
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (M.W.); (H.L.); (Y.W.); (M.S.)
- The Provincial and Ministerial Co-Founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Linzhi 860000, China
- Key Laboratory for the Genetic Improvement and Reproduction Technology of the Tibetan Swine, Linzhi 860000, China
| | - Yu Wang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (M.W.); (H.L.); (Y.W.); (M.S.)
- The Provincial and Ministerial Co-Founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Linzhi 860000, China
- Key Laboratory for the Genetic Improvement and Reproduction Technology of the Tibetan Swine, Linzhi 860000, China
| | - Mingyang Sun
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (M.W.); (H.L.); (Y.W.); (M.S.)
- The Provincial and Ministerial Co-Founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Linzhi 860000, China
- Key Laboratory for the Genetic Improvement and Reproduction Technology of the Tibetan Swine, Linzhi 860000, China
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (M.W.); (H.L.); (Y.W.); (M.S.)
- The Provincial and Ministerial Co-Founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Linzhi 860000, China
- Key Laboratory for the Genetic Improvement and Reproduction Technology of the Tibetan Swine, Linzhi 860000, China
| |
Collapse
|
9
|
Hurley LL, Ton R, Rowe M, Buchanan KL, Griffith SC, Crino OL. Longitudinal covariation of testosterone and sperm quality across reproductive stages in the zebra finch. Horm Behav 2023; 153:105388. [PMID: 37276837 DOI: 10.1016/j.yhbeh.2023.105388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
Abstract
Birds that breed opportunistically maintain partial activation of reproductive systems to rapidly exploit environmental conditions when they become suitable for breeding. Maintaining reproductive systems outside of a breeding context is costly. For males, these costs are thought to include continual exposure to testosterone. Males of seasonally breeding birds minimise these costs by downregulating testosterone production outside of a breeding context. Opportunistically breeding birds trade off the need to rapidly initiate reproduction with the costs of elevated testosterone production. One way opportunistically breeding males could minimise these costs is through fine scale changes in testosterone production across discrete reproductive stages which have a greater or lesser requirement for active sperm production. Although spermatogenesis broadly depends on testosterone production, whether changes in testosterone levels across the reproductive stages affect sperm quality and production is unknown. Here, we measured testosterone, sperm quality, and body condition in male zebra finches at discrete stages within reproductive bouts (egg laying, incubation, nestling provisioning, and fledging) and across two consecutive reproductive events in captive male zebra finches (Taeniopygia castanotis). We also examined associations between male testosterone, sperm quality/production, body condition, and nestling body condition. We found that testosterone levels varied across discrete reproductive stages with the lowest levels during incubation and the highest following chick fledging. Testosterone levels were positively associated with sperm velocity and the proportion of motile sperm but were not associated with male body condition. We found no associations between paternal body condition, testosterone levels, or sperm traits with nestling body condition (a proxy for the reproductive quality of a male and his partner). This study is the first to show that opportunistically breeding males vary testosterone synthesis and sperm traits at discrete stages within a reproductive event.
Collapse
Affiliation(s)
- Laura L Hurley
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Riccardo Ton
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Melissah Rowe
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), 6700 AB Wageningen, the Netherlands
| | - Katherine L Buchanan
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| | - Simon C Griffith
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ondi L Crino
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia; Division of Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, ACT 2600, Australia; College of Science and Engineering, Flinders University, Adelaide, SA 5042, Australia.
| |
Collapse
|
10
|
Zhang MF, Wan SC, Chen WB, Yang DH, Liu WQ, Li BL, Aierken A, Du XM, Li YX, Wu WP, Yang XC, Wei YD, Li N, Peng S, Li XL, Li GP, Hua JL. Transcription factor Dmrt1 triggers the SPRY1-NF-κB pathway to maintain testicular immune homeostasis and male fertility. Zool Res 2023; 44:505-521. [PMID: 37070575 PMCID: PMC10236308 DOI: 10.24272/j.issn.2095-8137.2022.440] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
Bacterial or viral infections, such as Brucella, mumps virus, herpes simplex virus, and Zika virus, destroy immune homeostasis of the testes, leading to spermatogenesis disorder and infertility. Of note, recent research shows that SARS-CoV-2 can infect male gonads and destroy Sertoli and Leydig cells, leading to male reproductive dysfunction. Due to the many side effects associated with antibiotic therapy, finding alternative treatments for inflammatory injury remains critical. Here, we found that Dmrt1 plays an important role in regulating testicular immune homeostasis. Knockdown of Dmrt1 in male mice inhibited spermatogenesis with a broad inflammatory response in seminiferous tubules and led to the loss of spermatogenic epithelial cells. Chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) revealed that Dmrt1 positively regulated the expression of Spry1, an inhibitory protein of the receptor tyrosine kinase (RTK) signaling pathway. Furthermore, immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP) analysis indicated that SPRY1 binds to nuclear factor kappa B1 (NF-κB1) to prevent nuclear translocation of p65, inhibit activation of NF-κB signaling, prevent excessive inflammatory reaction in the testis, and protect the integrity of the blood-testis barrier. In view of this newly identified Dmrt1- Spry1-NF-κB axis mechanism in the regulation of testicular immune homeostasis, our study opens new avenues for the prevention and treatment of male reproductive diseases in humans and livestock.
Collapse
Affiliation(s)
- Meng-Fei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shi-Cheng Wan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Bo Chen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Dong-Hui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Qing Liu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- Center of Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam 1105AZ, Amsterdam, Netherlands
| | - Ba-Lun Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Aili Aierken
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiao-Min Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yun-Xiang Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Ping Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xin-Chun Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yu-Dong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xue-Ling Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Guang-Peng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Jin-Lian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
11
|
Hazim Hamoode R, A. Alkubaisy S, A. Sattar D, S. Hamzah S, Hadi Saleh T, Laftaah Al-Rubaii BA. Detection of anti-testicular antibodies among infertile males using indirect immunofluorescent technique. Biomedicine (Taipei) 2022. [DOI: 10.51248/.v42i5.1963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Introduction and Aim: The antinuclear antibodies (ANA) are unwanted molecules which bind and destroy certain structures within the nucleus. Immunofluorescence is a powerful technique that utilizes fluorescent-labeled antibodies to detect specific target antigens. The aim of this study was to detect the anti-testicular antibody among infertile males in Baghdad city and determine the most common type of infertility.
Materials and Methods: The study involved 73 male (53 infertile and 20 non-infertile) volunteers, at Kamal Al-Samarrai Teaching Hospital in Baghdad, Iraq. Serum collected from the study subjects was tested for steroid-cell antibodies (STC-Ab), anti-nuclear antibody (ANA) and anti-testicular antibodies (ATCA) by Indirect Immunofluorescence assay (IIFA). Data obtained was subjected to statistical analysis using the SPSS program.
Results: In the current study 52.9% of infertile men tested positive for testicular antibodies compared to the control group. The highest rate of testicular cell antibodies was observed in the serum of infertile patients aged between 30-39 years and the lowest in patients aged 50-59 years. The marriage duration among those showing the highest percentage of antibodies against testicular cells was 1-10 years. Study for the association of male infertility type to those positive for ATCA showed almost all (85.7%) patients with necrozoospermia to be positive for ATCA. This was followed by males with azoospermia (50%) and oligospermia (46.9%). The patients were negative for Addison’s disease while a few (28.6%) were positive for lupus erythematosus.
Collapse
|
12
|
O'Donnell L, Smith LB, Rebourcet D. Sperm-specific proteins: new implications for diagnostic development and cancer immunotherapy. Curr Opin Cell Biol 2022; 77:102104. [PMID: 35671587 DOI: 10.1016/j.ceb.2022.102104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Spermatozoa are comprised of many unique proteins not expressed elsewhere. Sperm-specific proteins are first expressed at puberty, after the development of immune tolerance to self-antigens, and have been assumed to remain confined inside the seminiferous tubules, protected from immune cell recognition by various mechanisms of testicular immune privilege. However, new data has shown that sperm-specific proteins are released by the tubules into the surrounding interstitial fluid; from here they can contact immune cells, potentially promote immune tolerance, and enter the circulation. These new findings have clinical implications for diagnostics and therapeutics targeted at a specific class of proteins known as cancer-testis antigens (CTA), the opportunity to identify new communication pathways in the testis, and to discover new ways to monitor testis function.
Collapse
Affiliation(s)
- Liza O'Donnell
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, 3168, Victoria, Australia; Monash University, Clayton, 3168, Victoria, Australia.
| | - Lee B Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Griffith University, Parklands Drive, Southport, 4222, Queensland, Australia
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
13
|
Real FM, Lao-Pérez M, Burgos M, Mundlos S, Lupiáñez DG, Jiménez R, Barrionuevo FJ. Cell adhesion and immune response, two main functions altered in the transcriptome of seasonally regressed testes of two mammalian species. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2022; 340:231-244. [PMID: 35535962 DOI: 10.1002/jez.b.23142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/22/2022] [Accepted: 04/09/2022] [Indexed: 12/13/2022]
Abstract
In species with seasonal breeding, male specimens undergo substantial testicular regression during the nonbreeding period of the year. However, the molecular mechanisms that control this biological process are largely unknown. Here, we report a transcriptomic analysis on the Iberian mole, Talpa occidentalis, in which the desquamation of live, nonapoptotic germ cells is the major cellular event responsible for testis regression. By comparing testes at different reproductive states (active, regressing, and inactive), we demonstrate that the molecular pathways controlling the cell adhesion function in the seminiferous epithelium, such as the MAPK, ERK, and TGF-β signaling, are altered during the regression process. In addition, inactive testes display a global upregulation of genes associated with immune response, indicating a selective loss of the "immune privilege" that normally operates in sexually active testes. Interspecies comparative analyses using analogous data from the Mediterranean pine vole, a rodent species where testis regression is controlled by halting meiosis entry, revealed a common gene expression signature in the regressed testes of these two evolutionary distant species. Our study advances in the knowledge of the molecular mechanisms associated to gonadal seasonal breeding, highlighting the existence of a conserved transcriptional program of testis involution across mammalian clades.
Collapse
Affiliation(s)
- Francisca M Real
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Miguel Lao-Pérez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Stefan Mundlos
- RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Darío G Lupiáñez
- Epigenetics and Sex Development Group, Max-Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Francisco J Barrionuevo
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| |
Collapse
|
14
|
Hawthorne B, Lund K, Freggiaro S, Kaga R, Meng J. The mechanism of the cytotoxic effect of Panax notoginseng extracts on prostate cancer cells. Biomed Pharmacother 2022; 149:112887. [PMID: 35367754 DOI: 10.1016/j.biopha.2022.112887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
INTRODUCTION Panax notoginseng (Burkill) F.H. commonly referred to as Sanqi, is a Chinese herb that has long been used to treat various conditions including blood disorders and cardiovascular diseases. While Panax notoginseng has been used as an anti-cancer medicinal herb in recent years, how it achieves this therapeutic effect has not been thoroughly elucidated. The purpose of this study was to reveal more about the mechanism of the cytotoxic effect of Panax notoginseng on prostate cancer (PCa) cells. METHODS Ethanol extract of Panax notoginseng root was authenticated using high-performance liquid chromatography (HPLC). The cytotoxic activity of this herb against PCa cells was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method, flow cytometry, and enzyme-linked immunosorbent assay (ELISA). RESULTS The assessment of cellular metabolic activity demonstrated that Panax notoginseng reduces the viability of LNCaP and 22Rv1 cells in a dose-dependent manner. Annexin-V binding flow cytometry assay showed that Panax notoginseng induces apoptosis in PCa cells. Cell cycle analysis by quantification of DNA content using flow cytometry showed that Panax notoginseng arrests the cell cycle at the G2/M phase in both LNCaP and 22Rv1 cells. Moreover, ELISA demonstrated that Panax notoginseng-treated PCa cells secrete significantly less tumor-promoting cytokine interleukin-4 (IL-4) to the supernatant compared with controls. CONCLUSIONS These results provide evidence for the cytotoxic effects of Panax notoginseng on PCa cell lines. This botanical is a promising candidate for the complementary and integrative medicine treatment of PCa and further studies are indicated to determine the anti-cancer mechanism of Panax notoginseng.
Collapse
Affiliation(s)
- Benjamin Hawthorne
- School of Naturopathic Medicine, Bastyr University, 14500 Juanita Drive NE, Kenmore, WA 98028, USA
| | - Kaleb Lund
- School of Naturopathic Medicine, Bastyr University, 14500 Juanita Drive NE, Kenmore, WA 98028, USA
| | - Sydney Freggiaro
- School of Naturopathic Medicine, Bastyr University, 14500 Juanita Drive NE, Kenmore, WA 98028, USA
| | - Risa Kaga
- School of Natural Health Arts & Sciences, Bastyr University, 14500 Juanita Drive NE, Kenmore, WA 98028, USA
| | - Jing Meng
- School of Natural Health Arts & Sciences, Bastyr University, 14500 Juanita Drive NE, Kenmore, WA 98028, USA.
| |
Collapse
|
15
|
Adamczewska D, Słowikowska-Hilczer J, Walczak-Jędrzejowska R. The Fate of Leydig Cells in Men with Spermatogenic Failure. Life (Basel) 2022; 12:570. [PMID: 35455061 PMCID: PMC9028943 DOI: 10.3390/life12040570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/25/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
The steroidogenic cells in the testicle, Leydig cells, located in the interstitial compartment, play a vital role in male reproductive tract development, maintenance of proper spermatogenesis, and overall male reproductive function. Therefore, their dysfunction can lead to all sorts of testicular pathologies. Spermatogenesis failure, manifested as azoospermia, is often associated with defective Leydig cell activity. Spermatogenic failure is the most severe form of male infertility, caused by disorders of the testicular parenchyma or testicular hormone imbalance. This review covers current progress in knowledge on Leydig cells origin, structure, and function, and focuses on recent advances in understanding how Leydig cells contribute to the impairment of spermatogenesis.
Collapse
Affiliation(s)
| | | | - Renata Walczak-Jędrzejowska
- Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, 92-213 Lodz, Poland; (D.A.); (J.S.-H.)
| |
Collapse
|
16
|
Wang JM, Li ZF, Yang WX. What Does Androgen Receptor Signaling Pathway in Sertoli Cells During Normal Spermatogenesis Tell Us? Front Endocrinol (Lausanne) 2022; 13:838858. [PMID: 35282467 PMCID: PMC8908322 DOI: 10.3389/fendo.2022.838858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/01/2022] [Indexed: 01/18/2023] Open
Abstract
Androgen receptor signaling pathway is necessary to complete spermatogenesis in testes. Difference between androgen binding location in Sertoli cell classifies androgen receptor signaling pathway into classical signaling pathway and non-classical signaling pathway. As the only somatic cell type in seminiferous tubule, Sertoli cells are under androgen receptor signaling pathway regulation via androgen receptor located in cytoplasm and plasma membrane. Androgen receptor signaling pathway is able to regulate biological processes in Sertoli cells as well as germ cells surrounded between Sertoli cells. Our review will summarize the major discoveries of androgen receptor signaling pathway in Sertoli cells and the paracrine action on germ cells. Androgen receptor signaling pathway regulates Sertoli cell proliferation and maturation, as well as maintain the integrity of blood-testis barrier formed between Sertoli cells. Also, Spermatogonia stem cells achieve a balance between self-renewal and differentiation under androgen receptor signaling regulation. Meiotic and post-meiotic processes including Sertoli cell - Spermatid attachment and Spermatid development are guaranteed by androgen receptor signaling until the final sperm release. This review also includes one disease related to androgen receptor signaling dysfunction named as androgen insensitivity syndrome. As a step further ahead, this review may be conducive to develop therapies which can cure impaired androgen receptor signaling in Sertoli cells.
Collapse
|
17
|
Liu N, Qadri F, Busch H, Huegel S, Sihn G, Chuykin I, Hartmann E, Bader M, Rother F. Kpna6 deficiency causes infertility in male mice by disrupting spermatogenesis. Development 2021; 148:272018. [PMID: 34473250 PMCID: PMC8513612 DOI: 10.1242/dev.198374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 08/18/2021] [Indexed: 11/20/2022]
Abstract
Spermatogenesis is driven by an ordered series of events, which rely on trafficking of specific proteins between nucleus and cytoplasm. The karyopherin α family of proteins mediates movement of specific cargo proteins when bound to karyopherin β. Karyopherin α genes have distinct expression patterns in mouse testis, implying they may have unique roles during mammalian spermatogenesis. Here, we use a loss-of-function approach to determine specifically the role of Kpna6 in spermatogenesis and male fertility. We show that ablation of Kpna6 in male mice leads to infertility and has multiple cumulative effects on both germ cells and Sertoli cells. Kpna6-deficient mice exhibit impaired Sertoli cell function, including loss of Sertoli cells and a compromised nuclear localization of the androgen receptor. Furthermore, our data demonstrate devastating defects on spermiogenesis, including incomplete sperm maturation and a massive reduction in sperm number, accompanied by disturbed histone-protamine exchange, differential localization of the transcriptional regulator BRWD1 and altered expression of RFX2 target genes. Our work uncovers an essential role of Kpna6 in spermatogenesis and, hence, in male fertility. Summary: Two different mouse models delineate the morphological and functional impact of Kpna6 on spermatogenesis and Sertoli cell function and show that this protein is crucial for fertility in male mice.
Collapse
Affiliation(s)
- Na Liu
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | | | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Lübeck 23562, Germany
| | - Stefanie Huegel
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Gabin Sihn
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Ilya Chuykin
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Enno Hartmann
- Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Franziska Rother
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| |
Collapse
|
18
|
Fang Y, Su Y, Xu J, Hu Z, Zhao K, Liu C, Zhang H. Varicocele-Mediated Male Infertility: From the Perspective of Testicular Immunity and Inflammation. Front Immunol 2021; 12:729539. [PMID: 34531872 PMCID: PMC8438154 DOI: 10.3389/fimmu.2021.729539] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/16/2021] [Indexed: 01/14/2023] Open
Abstract
Background Varicocele (VC) is present in 35 - 40% of men with infertility. However, current surgical and antioxidant treatments are not completely effective. In addition to oxidative stress, it is likely that other factors such as testicular immune microenvironment disorder contribute to irreversible testicular. Evidence suggests that VC is associated with anti-sperm antibodies (ASAs), spermatogenesis and testosterone secretion abnormalities, and testicular cytokine production. Moreover, inhibition of inflammation can alleviate VC-mediated pathogenesis. The normal function of the testis depends on its immune tolerance mechanism. Testicular immune regulation is complex, and many infectious or non-infectious diseases may damage this precision system. Results The testicular immune microenvironment is composed of common immune cells and other cells involved in testicular immunity. The former includes testicular macrophages, T cells, dendritic cells (DCs), and mast cells, whereas the latter include Leydig cells and Sertoli cells (SCs). In animal models and in patients with VC, most studies have revealed an abnormal increase in the levels of ASAs and pro-inflammatory cytokines such as interleukin (IL)-1 and tumor necrosis factor (TNF)-alpha in the seminal plasma, testicular tissue, and even peripheral blood. It is also involved in the activation of potential inflammatory pathways, such as the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing (NLRP)-3 pathway. Finally, the development of VC-mediated infertility (VMI) may be facilitated by abnormal permeability of proteins, such as claudin-11, that constitute the blood-testis barrier (BTB). Conclusions The testicular immune response, including the production of ASAs and inflammatory factors, activation of inflammatory pathways, and destruction of the BTB may be involved in the pathogenesis of VMI it is necessary to further explore how patient outcomes can be improved through immunotherapy.
Collapse
Affiliation(s)
- Yiwei Fang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Hu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Ye L, Huang W, Liu S, Cai S, Hong L, Xiao W, Thiele K, Zeng Y, Song M, Diao L. Impacts of Immunometabolism on Male Reproduction. Front Immunol 2021; 12:658432. [PMID: 34367130 PMCID: PMC8334851 DOI: 10.3389/fimmu.2021.658432] [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: 01/25/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The physiological process of male reproduction relies on the orchestration of neuroendocrine, immune, and energy metabolism. Spermatogenesis is controlled by the hypothalamic-pituitary-testicular (HPT) axis, which modulates the production of gonadal steroid hormones in the testes. The immune cells and cytokines in testes provide a protective microenvironment for the development and maturation of germ cells. The metabolic cellular responses and processes in testes provide energy production and biosynthetic precursors to regulate germ cell development and control testicular immunity and inflammation. The metabolism of immune cells is crucial for both inflammatory and anti-inflammatory responses, which supposes to affect the spermatogenesis in testes. In this review, the role of immunometabolism in male reproduction will be highlighted. Obesity, metabolic dysfunction, such as type 2 diabetes mellitus, are well documented to impact male fertility; thus, their impacts on the immune cells distributed in testes will also be discussed. Finally, the potential significance of the medicine targeting the specific metabolic intermediates or immune metabolism checkpoints to improve male reproduction will also be reassessed.
Collapse
Affiliation(s)
- Lijun Ye
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Wensi Huang
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Su Liu
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Songchen Cai
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Ling Hong
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Weiqiang Xiao
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Mingzhe Song
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
20
|
O'Donnell L, Smith LB, Rebourcet D. Sertoli cells as key drivers of testis function. Semin Cell Dev Biol 2021; 121:2-9. [PMID: 34229950 DOI: 10.1016/j.semcdb.2021.06.016] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022]
Abstract
Sertoli cells are the orchestrators of spermatogenesis; they support fetal germ cell commitment to the male pathway and are essential for germ cell development, from maintenance of the spermatogonial stem cell niche and spermatogonial populations, through meiosis and spermiogeneis and to the final release of mature spermatids during spermiation. However, Sertoli cells are also emerging as key regulators of other testis somatic cells, including supporting peritubular myoid cell development in the pre-pubertal testis and supporting the function of the testicular vasculature and in contributing to testicular immune privilege. Sertoli cells also have a major role in regulating androgen production within the testis, by specifying interstitial cells to a steroidogenic fate, contributing to androgen production in the fetal testis, and supporting fetal and adult Leydig cell development and function. Here, we provide an overview of the specific roles for Sertoli cells in the testis and highlight how these cells are key drivers of testicular sperm output, and of adult testis size and optimal function of other testicular somatic cells, including the steroidogenic Leydig cells.
Collapse
Affiliation(s)
- Liza O'Donnell
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; Monash University, Clayton 3168, Victoria, Australia.
| | - Lee B Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
21
|
Zheng W, Zhang S, Jiang S, Huang Z, Chen X, Guo H, Li M, Zheng S. Evaluation of immune status in testis and macrophage polarization associated with testicular damage in patients with nonobstructive azoospermia. Am J Reprod Immunol 2021; 86:e13481. [PMID: 34192390 DOI: 10.1111/aji.13481] [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: 04/04/2021] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Immune cells residing in the testicular interstitial space form the immunological microenvironment of the testis. They are assumed to play a role in maintaining testicular homeostasis and immune privilege. However, the immune status and related cell polarization in patients with nonobstructive azoospermia (NOA) remains poorly characterized. System evaluation of the testis immunological microenvironment in NOA patients may help to reveal the mechanisms of idiopathic azoospermia. STUDY DESIGN The gene expression patterns of immune cells in normal human testes were systematically analyzed by single-cell RNA sequencing (scRNA-seq) and preliminarily verification by the human protein atlas (HPA) online database. The immune cell infiltration profiles and immune status of patients with NOA was analyzed by single-sample gene set enrichment analysis (ssGSEA) and gene set variation analysis (GSVA) based on four independent public microarray datasets (GSE45885, GSE45887, GSE9210, and GSE145467), obtained from Gene Expression Omnibus (GEO) online database. The relationship between immune cells and spermatogenesis score was further analyzed by Spearman correlation analysis. Finally, immunohistochemistry (IHC) staining was performed to identify the main immune cell types and their polarization status in patients with NOA. RESULTS Both scRNA-seq and HPA analysis showed that testicular macrophages represent the largest pool of immune cells in the normal testis, and also exhibit an attenuated inflammatory response by expressing high levels of tolerance proteins (CD163, IL-10, TGF-β, and VEGF) and reduced expression of TLR signaling pathway-related genes. Correlation analysis revealed that the testicular immune score and macrophages including M1 and M2 macrophages were significantly negatively correlated with spermatogenesis score in patients with NOA (GSE45885 and GSE45887). In addition, the number of M1 and M2 macrophages was significantly higher in patients with NOA (GSE9210 and GSE145467) than in normal testis. GSVA analysis indicated that the immunological microenvironment in NOA tissues was manifested by activated immune system and pro-inflammatory status. IHC staining results showed that the number of M1 and M2 macrophages was significantly higher in NOA tissues than in normal testis and negatively correlated with the Johnson score. CONCLUSION Testicular macrophage polarization may play a vital role in NOA development and is a promising potential therapeutic target.
Collapse
Affiliation(s)
- Wenzhong Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shaoqin Jiang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhangcheng Huang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaobao Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Mengqiang Li
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
22
|
Li T, Wang X, Luo R, An X, Zhang Y, Zhao X, Ma Y. Integrating miRNA and mRNA Profiling to Assess the Potential miRNA-mRNA Modules Linked With Testicular Immune Homeostasis in Sheep. Front Vet Sci 2021; 8:647153. [PMID: 34113669 PMCID: PMC8185144 DOI: 10.3389/fvets.2021.647153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/07/2021] [Indexed: 12/05/2022] Open
Abstract
Beyond its well-known role in spermatogenesis and androgen production, mammalian testes are increasingly recognized as an immune-privileged organ for protecting autoantigenic germ cells, especially meiotic and postmeiotic germ cells, from systemic immune responses. Despite its importance, the molecular mechanisms underlying this regulation in mammals, including sheep, are far from known. In this study, we searched for the genes associated with testicular immune privilege and assessed their possible modulating mechanisms by analyzing systematic profiling of mRNAs and miRNAs on testicular tissues derived from prepubertal and postpubertal Tibetan sheep acquired by RNA sequencing. We identified 1,118 differentially expressed (DE) mRNAs associated with immunity (245 increased mRNAs and 873 decreased mRNAs) and 715 DE miRNAs (561 increased miRNAs and 154 decreased miRNAs) in postpubertal testes compared with prepuberty. qPCR validations for 20 DE mRNAs and 16 miRNAs showed that the RNA-seq results are reliable. By using Western blot, the postpubertal testes exhibited decreased protein abundance of CD19 and TGFBR2 (two proteins encoded by DE mRNAs) when compared with prepuberty, consistent with mRNA levels. The subsequent immunofluorescent staining showed that the positive signals for the CD19 protein were observed mainly in Sertoli cells and the basement membrane of pre- and postpubertal testes, as well as the prepubertal testicular vascular endothelium. The TGFBR2 protein was found mostly in interstitial cells and germ cells of pre- and postpubertal testes. Functional enrichment analysis indicated that DE mRNAs were mainly enriched in biological processes or pathways strongly associated with the blood–testis barrier (BTB) function. Many decreased mRNAs with low expression abundance were significantly enriched in pathways related to immune response. Also, multiple key miRNA-target negative correlation regulatory networks were subsequently established. Furthermore, we verified the target associations between either oar-miR-29b or oar-miR-1185-3p and ITGB1 by dual-luciferase reporter assay. Finally, a putative schematic model of the miRNA-mRNA-pathway network mediated by immune homeostasis-related genes was proposed to show their potential regulatory roles in sheep testicular privilege. Taken together, we conclude that many immune-related genes identified in this study are negatively regulated by potential miRNAs to participate in the homeostatic regulation of testicular immune privilege of sheep by sustaining BTB function and inhibiting immune responses under normal physiological conditions. This work offers the first global view of the expression profiles of miRNAs/mRNAs involved in sheep testicular immune privilege and how the genes potentially contribute to immune-homeostatic maintenance.
Collapse
Affiliation(s)
- Taotao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China.,Sheep Breeding Biotechnology Engineering Laboratory of Gansu Province, Minqin, China
| | - Xia Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Ruirui Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xuejiao An
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Youji Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China.,Sheep Breeding Biotechnology Engineering Laboratory of Gansu Province, Minqin, China
| |
Collapse
|
23
|
Li T, Luo R, Wang X, Wang H, Zhao X, Guo Y, Jiang H, Ma Y. Unraveling Stage-Dependent Expression Patterns of Circular RNAs and Their Related ceRNA Modulation in Ovine Postnatal Testis Development. Front Cell Dev Biol 2021; 9:627439. [PMID: 33816472 PMCID: PMC8017185 DOI: 10.3389/fcell.2021.627439] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) have been shown to function in the reproductive systems including testis. However, their expression, as well as function in testicular development of sheep remain undefined. Herein, we performed RNA sequencing to reveal circRNA temporal expression patterns in testes of Tibetan sheep from different stages of maturation (3M, 3-month-old; 1Y, 1-year-old; 3Y, 3-year-old). A total of 3,982, 414, and 4,060 differentially expressed (DE) circRNAs were uncovered from 3M vs 1Y, 1Y vs 3Y, and 3M vs 3Y, respectively. Functional enrichment assessment indicated that the source genes of DE circRNAs were primarily engaged in spermatogenesis and testicular immune privilege including blood-testis barrier (BTB). We subsequently constructed the core circRNA-miRNA-mRNA interaction network for genes related to testicular function, such as spermatogenesis, germ cell development, BTB, and cell cycle/meiosis. Furthermore, we validated the target associations between either circ_024949, circ_026259 or IGF1, and oar-miR-29b in this network, and revealed their similar expression signatures in developmental testes that they were extensively expressed in germ cells, Leydig cells, and Sertoli cells, thus suggesting their broad functions in the functional maintenance of Leydig cells and Sertoli cells, as well as the development and maturation of male germ cells. Meanwhile, circ_026259 was shown to promote IGF1 expression through inhibition of oar-miR-29b in sheep Sertoli cells. This work gives the first global view for the expression and regulation of circRNAs in sheep testis, which will be helpful for providing new insights into the molecular mechanism of ovine testis function.
Collapse
Affiliation(s)
- Taotao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Ruirui Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xia Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Huihui Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yunxia Guo
- College of Life Science, Hebei Agricultural University, Baoding, China
| | - Haitao Jiang
- Wenshang County Inspection and Testing Center, Jining, China
| | - Youji Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
24
|
Abstract
Puberty is characterized by major changes in the anatomy and function of reproductive organs. Androgen activity is low before puberty, but during pubertal development, the testes resume the production of androgens. Major physiological changes occur in the testicular cell compartments in response to the increase in intratesticular testosterone concentrations and androgen receptor expression. Androgen activity also impacts on the internal and external genitalia. In target cells, androgens signal through a classical and a nonclassical pathway. This review addresses the most recent advances in the knowledge of the role of androgen signaling in postnatal male sexual development, with a special emphasis on human puberty.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
25
|
Massoud D, Lao-Pérez M, Ortega E, Burgos M, Jiménez R, Barrionuevo FJ. Divergent Seasonal Reproductive Patterns in Syntopic Populations of Two Murine Species in Southern Spain, Mus spretus and Apodemus sylvaticus. Animals (Basel) 2021; 11:ani11020243. [PMID: 33498171 PMCID: PMC7908971 DOI: 10.3390/ani11020243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/16/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary In temperate zones of the Earth, most species reproduce in seasons providing the most favourable environmental conditions. Producing gametes is expensive in energetical terms, so both males and females either reduce or abolish gametogenesis during the non-breeding period. We thoroughly studied the testes of sexually inactive males of two rodents, the wood mouse, Apodemus sylvaticus, and the Algerian mouse, Mus spretus, in southern Iberian peninsula. These populations are syntopic, that is, animals of the two species share their territories and resources, so one would expect them to show similar or identical seasonal reproduction patterns. Contrarily, we found that both species reproduce during most of the year, but wood mice stop breeding in the summer whereas Algerian mice do it in winter. These divergent seasonal breeding patterns imply that either very subtle animal features and/or environmental cues operate to determine reproduction timing and support the notion that multiple models of circannual reproduction patterns are possible for different populations of the same species, showing that the mechanisms controlling seasonal reproduction are in fact very plastic and fast evolving. Hence, small mammals probably have multiple ways available to get adapted to the unstable environmental conditions derived from the ongoing global climate change. Abstract In most mammals with seasonal reproduction, males undergo testis regression during the non-breeding period. We performed a morphological, hormonal, functional, and molecular study of the testes of sexually inactive males of two species of murine rodents, the wood mouse, Apodemus sylvaticus, and the Algerian mouse, Mus spretus, in syntopic populations of southern Iberian peninsula. Both species reproduce during most of the year, but wood mice stop breeding in the summer whereas Algerian mice do it in winter. Sexually inactive males of A. sylvaticus show complete testis regression with reduced levels of serum testosterone and abnormal distribution of cell-adhesion molecules. Contrarily, inactive males of M. spretus maintain almost normal spermotogenesis despite a significant reduction of androgenic function. The lack of an evident explanation for the divergent seasonal breeding patterns found in southern populations of A. sylvaticus and M. spretus, compared with northern ones, implies that very subtle species/population-specific features and/or non-conspicuous environmental cues probably operate to determine their seasonal breeding pattern. These results also support the notion that multiple models of circannual testis variation are possible for different populations of the same species, showing that the mechanisms controlling seasonal reproduction are in fact very plastic and fast evolving.
Collapse
Affiliation(s)
- Diaa Massoud
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Avenida del Conocimiento S/N, 18016 Armilla, Granada, Spain; (D.M.); (M.L.-P.); (M.B.); (F.J.B.)
- Department of Zoology, Faculty of Science, Fayoum University, Gamma St., Keman Square, Fayoum 63514, Egypt
| | - Miguel Lao-Pérez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Avenida del Conocimiento S/N, 18016 Armilla, Granada, Spain; (D.M.); (M.L.-P.); (M.B.); (F.J.B.)
| | - Esperanza Ortega
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Avenida de la Investigación 11, 18071 Granada, Spain;
| | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Avenida del Conocimiento S/N, 18016 Armilla, Granada, Spain; (D.M.); (M.L.-P.); (M.B.); (F.J.B.)
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Avenida del Conocimiento S/N, 18016 Armilla, Granada, Spain; (D.M.); (M.L.-P.); (M.B.); (F.J.B.)
- Correspondence:
| | - Francisco J. Barrionuevo
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Avenida del Conocimiento S/N, 18016 Armilla, Granada, Spain; (D.M.); (M.L.-P.); (M.B.); (F.J.B.)
| |
Collapse
|
26
|
Souza TL, Batschauer AR, Brito PM, Martino-Andrade AJ, Ortolani-Machado CF. Evaluation of testicular structure in mice after exposure to environmentally relevant doses of manganese during critical windows of development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 207:111537. [PMID: 33254399 DOI: 10.1016/j.ecoenv.2020.111537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/29/2020] [Accepted: 10/16/2020] [Indexed: 06/12/2023]
Abstract
Despite being an essential trace element with great importance for vital metabolic activities, the manganese (Mn) can also cause damage to organ systems. However, data on the effect of this metal on the male reproductive system are limited, especially using relevant doses to human exposure. The present study aimed to evaluate and compare the effects of Mn exposure on the testicular structure of mice. Three experiments were conducted: (I) direct exposure to realistic doses (0.013, 0.13, and 1.3 mg/kg/day of MnCl2); (II) parental and direct exposure to realistic doses (as in experiment I), where the animals were exposed during intrauterine development and from lactation until reproductive maturity; (III) direct exposure to high doses (15, 30, and 60 mg/kg/day of MnCl2). Biometric, histopathological, histomorphometric and stereological parameters of the testis were evaluated, in addition to sperm morphology. Bioinformatic analyses were performed to identify potential Mn binding sites in 3β-HSD and P450ssc, as well as their protein-protein interaction network. The results obtained were compared using the integrated biomarker response index (IBR). There was an increase of seminiferous tubules pathologies in all experimental conditions tested, with effects on tubular volume, as well as a reduction in tubular diameter. The IBR analyses showed that parental and direct exposure had a significant negative effect on the testicular structure due to the exposure of this metal to sensitive periods of animal development. This study suggests that Mn has the potential to alter the morphological parameters of the testes, affecting the spermatogenesis in mice.
Collapse
Affiliation(s)
- Tugstênio L Souza
- Laboratory of Embryotoxicology, Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil.
| | - Amândia R Batschauer
- Laboratory of Embryotoxicology, Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Patricia M Brito
- Laboratory of Cell Toxicology, Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Anderson J Martino-Andrade
- Laboratory of Endocrine Physiology and Animal Reproduction, Department of Physiology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Claudia F Ortolani-Machado
- Laboratory of Embryotoxicology, Department of Cell Biology, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
27
|
Abstract
Mammalian spermatogenesis is a carefully orchestrated male germ cell differentiation process by which spermatogonia differentiate to spermatozoa in the testis. A highly organized testicular microenvironment is therefore necessary to support spermatogenesis. Regarding immunologic aspects, the testis adapts a specialized immune environment for the protection of male germ cells and testicular functions. The mammalian testis possesses two immunologic features: (1) it is an immunoprivileged organ where immunogenic germ cells do not induce deleterious immune responses under physiologic conditions; and (2) it creates its own effective innate defense system against microbial infection. Various pathologic conditions may disrupt testicular immune homeostasis, thereby resulting in a detrimental immune response and perturbing testicular functions, one of the etiologic factors of male infertility. Understanding the mechanisms underlying immunoregulation in the testis can aid in establishing strategies for the prevention and therapy of immunologic testicular dysfunction and male infertility. This chapter focuses on the mechanisms underlying immune privilege, local innate immunity, and immunologic diseases of the testis.
Collapse
|
28
|
Molecular insights into hormone regulation via signaling pathways in Sertoli cells: With discussion on infertility and testicular tumor. Gene 2020; 753:144812. [DOI: 10.1016/j.gene.2020.144812] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
|
29
|
A framework for high-resolution phenotyping of candidate male infertility mutants: from human to mouse. Hum Genet 2020; 140:155-182. [PMID: 32248361 DOI: 10.1007/s00439-020-02159-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022]
Abstract
Male infertility is a heterogeneous condition of largely unknown etiology that affects at least 7% of men worldwide. Classical genetic approaches and emerging next-generation sequencing studies support genetic variants as a frequent cause of male infertility. Meanwhile, the barriers to transmission of this disease mean that most individual genetic cases will be rare, but because of the large percentage of the genome required for spermatogenesis, the number of distinct causal mutations is potentially large. Identifying bona fide causes of male infertility thus requires advanced filtering techniques to select for high-probability candidates, including the ability to test causality in animal models. The mouse remains the gold standard for defining the genotype-phenotype connection in male fertility. Here, we present a best practice guide consisting of (a) major points to consider when interpreting next-generation sequencing data performed on infertile men, and, (b) a systematic strategy to categorize infertility types and how they relate to human male infertility. Phenotyping infertility in mice can involve investigating the function of multiple cell types across the testis and epididymis, as well as sperm function. These findings will feed into the diagnosis and treatment of male infertility as well as male health broadly.
Collapse
|
30
|
Le Tortorec A, Matusali G, Mahé D, Aubry F, Mazaud-Guittot S, Houzet L, Dejucq-Rainsford N. From Ancient to Emerging Infections: The Odyssey of Viruses in the Male Genital Tract. Physiol Rev 2020; 100:1349-1414. [PMID: 32031468 DOI: 10.1152/physrev.00021.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The male genital tract (MGT) is the target of a number of viral infections that can have deleterious consequences at the individual, offspring, and population levels. These consequences include infertility, cancers of male organs, transmission to the embryo/fetal development abnormalities, and sexual dissemination of major viral pathogens such as human immunodeficiency virus (HIV) and hepatitis B virus. Lately, two emerging viruses, Zika and Ebola, have additionally revealed that the human MGT can constitute a reservoir for viruses cleared from peripheral circulation by the immune system, leading to their sexual transmission by cured men. This represents a concern for future epidemics and further underlines the need for a better understanding of the interplay between viruses and the MGT. We review here how viruses, from ancient viruses that integrated the germline during evolution through old viruses (e.g., papillomaviruses originating from Neanderthals) and more modern sexually transmitted infections (e.g., simian zoonotic HIV) to emerging viruses (e.g., Ebola and Zika) take advantage of genital tract colonization for horizontal dissemination, viral persistence, vertical transmission, and endogenization. The MGT immune responses to viruses and the impact of these infections are discussed. We summarize the latest data regarding the sources of viruses in semen and the complex role of this body fluid in sexual transmission. Finally, we introduce key animal findings that are relevant for our understanding of viral infection and persistence in the human MGT and suggest future research directions.
Collapse
Affiliation(s)
- Anna Le Tortorec
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Giulia Matusali
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Dominique Mahé
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Florence Aubry
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Séverine Mazaud-Guittot
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Laurent Houzet
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| |
Collapse
|
31
|
Kalavska K, Kucerova L, Schmidtova S, Chovanec M, Mego M. Cancer Stem Cell Niche and Immune-Active Tumor Microenvironment in Testicular Germ Cell Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:111-121. [DOI: 10.1007/978-3-030-36214-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
32
|
Edelsztein NY, Rey RA. Importance of the Androgen Receptor Signaling in Gene Transactivation and Transrepression for Pubertal Maturation of the Testis. Cells 2019; 8:E861. [PMID: 31404977 PMCID: PMC6721648 DOI: 10.3390/cells8080861] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022] Open
Abstract
Androgens are key for pubertal development of the mammalian testis, a phenomenon that is tightly linked to Sertoli cell maturation. In this review, we discuss how androgen signaling affects Sertoli cell function and morphology by concomitantly inhibiting some processes and promoting others that contribute jointly to the completion of spermatogenesis. We focus on the molecular mechanisms that underlie anti-Müllerian hormone (AMH) inhibition by androgens at puberty, as well as on the role androgens have on Sertoli cell tight junction formation and maintenance and, consequently, on its effect on proper germ cell differentiation and meiotic onset during spermatogenesis.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina.
| |
Collapse
|
33
|
Griswold MD. 50 years of spermatogenesis: Sertoli cells and their interactions with germ cells. Biol Reprod 2018; 99:87-100. [PMID: 29462262 PMCID: PMC7328471 DOI: 10.1093/biolre/ioy027] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/22/2018] [Accepted: 02/02/2018] [Indexed: 01/15/2023] Open
Abstract
The complex morphology of the Sertoli cells and their interactions with germ cells has been a focus of investigators since they were first described by Enrico Sertoli. In the past 50 years, information on Sertoli cells has transcended morphology alone to become increasingly more focused on molecular questions. The goal of investigators has been to understand the role of the Sertoli cells in spermatogenesis and to apply that information to problems relating to male fertility. Sertoli cells are unique in that they are a nondividing cell population that is active for the reproductive lifetime of the animal and cyclically change morphology and gene expression. The numerous and distinctive junctional complexes and membrane specializations made by Sertoli cells provide a scaffold and environment for germ cell development. The increased focus of investigators on the molecular components and putative functions of testicular cells has resulted primarily from procedures that isolate specific cell types from the testicular milieu. Products of Sertoli cells that influence germ cell development and vice versa have been characterized from cultured cells and from the application of transgenic technologies. Germ cell transplantation has shown that the Sertoli cells respond to cues from germ cells with regard to developmental timing and has furthered a focus on spermatogenic stem cells and the stem cell niche. Very basic and universal features of spermatogenesis such as the cycle of the seminiferous epithelium and the spermatogenic wave are initiated by Sertoli cells and maintained by Sertoli-germ cell cooperation.
Collapse
Affiliation(s)
- Michael D Griswold
- Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
34
|
Fijak M, Pilatz A, Hedger MP, Nicolas N, Bhushan S, Michel V, Tung KSK, Schuppe HC, Meinhardt A. Infectious, inflammatory and 'autoimmune' male factor infertility: how do rodent models inform clinical practice? Hum Reprod Update 2018; 24:416-441. [PMID: 29648649 PMCID: PMC6016649 DOI: 10.1093/humupd/dmy009] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/02/2018] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Infection and inflammation of the reproductive tract are significant causes of male factor infertility. Ascending infections caused by sexually transmitted bacteria or urinary tract pathogens represent the most frequent aetiology of epididymo-orchitis, but viral, haematogenous dissemination is also a contributory factor. Limitations in adequate diagnosis and therapy reflect an obvious need for further understanding of human epididymal and testicular immunopathologies and their contribution to infertility. A major obstacle for advancing our knowledge is the limited access to suitable tissue samples. Similarly, the key events in the inflammatory or autoimmune pathologies affecting human male fertility are poorly amenable to close examination. Moreover, the disease processes generally have occurred long before the patient attends the clinic for fertility assessment. In this regard, data obtained from experimental animal models and respective comparative analyses have shown promise to overcome these restrictions in humans. OBJECTIVE AND RATIONALE This narrative review will focus on male fertility disturbances caused by infection and inflammation, and the usefulness of the most frequently applied animal models to study these conditions. SEARCH METHODS An extensive search in Medline database was performed without restrictions until January 2018 using the following search terms: 'infection' and/or 'inflammation' and 'testis' and/or 'epididymis', 'infection' and/or 'inflammation' and 'male genital tract', 'male infertility', 'orchitis', 'epididymitis', 'experimental autoimmune' and 'orchitis' or 'epididymitis' or 'epididymo-orchitis', antisperm antibodies', 'vasectomy'. In addition to that, reference lists of primary and review articles were reviewed for additional publications independently by each author. Selected articles were verified by each two separate authors and discrepancies discussed within the team. OUTCOMES There is clear evidence that models mimicking testicular and/or epididymal inflammation and infection have been instructive in a better understanding of the mechanisms of disease initiation and progression. In this regard, rodent models of acute bacterial epididymitis best reflect the clinical situation in terms of mimicking the infection pathway, pathogens selected and the damage, such as fibrotic transformation, observed. Similarly, animal models of acute testicular and epididymal inflammation using lipopolysaccharides show impairment of reproduction, endocrine function and histological tissue architecture, also seen in men. Autoimmune responses can be studied in models of experimental autoimmune orchitis (EAO) and vasectomy. In particular, the early stages of EAO development showing inflammatory responses in the form of peritubular lymphocytic infiltrates, thickening of the lamina propria of affected tubules, production of autoantibodies against testicular antigens or secretion of pro-inflammatory mediators, replicate observations in testicular sperm extraction samples of patients with 'mixed atrophy' of spermatogenesis. Vasectomy, in the form of sperm antibodies and chronic inflammation, can also be studied in animal models, providing valuable insights into the human response. WIDER IMPLICATIONS This is the first comprehensive review of rodent models of both infectious and autoimmune disease of testis/epididymis, and their clinical implications, i.e. their importance in understanding male infertility related to infectious and non-infectious/autoimmune disease of the reproductive organs.
Collapse
Affiliation(s)
- Monika Fijak
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Adrian Pilatz
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University of Giessen, Germany
| | - Mark P Hedger
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Nour Nicolas
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Vera Michel
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
| | - Kenneth S K Tung
- Departments of Pathology and Microbiology, Beirne Carter Center for Immunology Research, University of Virginia, 345 Crispell Drive, Charlottesville, VA, USA
| | - Hans-Christian Schuppe
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University of Giessen, Germany
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Aulweg 123, Giessen, Germany
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, Australia
| |
Collapse
|
35
|
Immune-Related Concepts in Biology and Treatment of Germ-Cell Tumors. Adv Urol 2018; 2018:3718165. [PMID: 29725351 PMCID: PMC5872660 DOI: 10.1155/2018/3718165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/22/2018] [Accepted: 02/04/2018] [Indexed: 12/22/2022] Open
Abstract
Germ-cell tumors (GCTs) are highly curable with chemotherapy. Salvage chemotherapy or surgery can cure a proportion of patients, but the ones failing these treatments will die of their disease in the young age. Immune checkpoint pathways are emerging as powerful targetable biomarkers, and a significant preclinical and clinical research is underway to widen our knowledge and expand the treatment possibilities with immune therapy. The concept of immune modulation that was currently adopted in many solid tumors is understudied in GCTs. Herein, we summarize the current knowledge of published literature discussing the immune mechanisms and immune therapy in GCTs.
Collapse
|
36
|
Sun Z, Nie Q, Zhang L, Niu R, Wang J, Wang S. Fluoride reduced the immune privileged function of mouse Sertoli cells via the regulation of Fas/FasL system. CHEMOSPHERE 2017; 168:318-325. [PMID: 27810530 DOI: 10.1016/j.chemosphere.2016.10.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/09/2016] [Accepted: 10/07/2016] [Indexed: 06/06/2023]
Abstract
Previous investigations have demonstrated the adverse impacts of fluoride on Sertoli cells (SCs), such as oxidative stress and apoptosis. SCs are the crucial cellular components that can create the immune privileged environment in testis. However, the effect of fluoride on SCs immune privilege is unknown. In this study, mouse SCs were exposed to sodium fluoride with varying concentrations of 10-5, 10-4, and 10-3 mol/L to establish the model of fluoride-treated SCs (F-SCs) in vitro. After 48 h of incubation, F-SCs were transplanted underneath the kidney capsule of mice for 21 days, or cocultured with spleen lymphocytes for another 48 h. Immunohistochemical analysis of GATA4 in SCs grafts underneath kidney capsule presented less SCs distribution and obvious immune cell infiltration in F-SCs groups. In addition, the levels of FasL protein and mRNA in non-cocultured F-SCs decreased with the increase of fluoride concentration. When cocultured with F-SCs, lymphocytes presented significantly high cell viability and low apoptosis in F-SCs groups. Protein and mRNA expressions of FasL in cocultured F-SCs and Fas in lymphocytes were reduced, and the caspase 8 and caspase 3 mRNA levels were also decreased in fluoride groups in a dose-dependent manner. These findings indicated that fluoride influenced the testicular immune privilege through disturbing the Fas/FasL system.
Collapse
Affiliation(s)
- Zilong Sun
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Qingli Nie
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Lianjie Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Ruiyan Niu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jundong Wang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China.
| | - Shaolin Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
37
|
Effect of extracellular matrix on testosterone production during in vitro culture of bovine testicular cells. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2017; 8:7-13. [PMID: 28473891 PMCID: PMC5413305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/05/2016] [Indexed: 11/06/2022]
Abstract
Testosterone is believed to play a significant role in spermatogenesis, but its contribution to the process of spermatogenesis is not completely understood. Given that extracellular matrix (ECM) facilitates differentiation of spermatogonial stem cells (SSCs) during culture, the present study was conducted to elucidate whether testosterone contribute to the permissive effect of ECM on SSCs differentiation. In experiment 1, testosterone production was measured in testicular cells cultured for 12 days on ECM or plastic (control). In experiment 2, testosterone production was assessed in testicular cells cultured on ECM or plastic (control) and exposed to different concentrations of hCG. In experiment 3, the gene expression of factors involved in testosterone production was analyzed. Testosterone concentration was lower in ECM than in the control group in experiment 1 (p < 0.05). In experiment 2, testosterone concentration was increased in response to hCG in both groups but cells cultured on ECM were more responsive to hCG than those cultured on plastic (p < 0.05). In the experiment 3, qRT-PCR revealed the inhibitory effect of ECM on the gene expression of steroidogenic acute regulatory protein (StAR) (p < 0.05). Nevertheless, the expression of LH receptor was greater in ECM-exposed than in unexposed cells (p < 0.05). In conclusion, the present study showed that inhibiting the expression of StAR, ECM could lower testosterone production by Leydig cells during in vitro culture. In addition, the results indicated that ECM could augment the responsiveness of Leydig cells to hCG through stimulating the expression of LH receptor.
Collapse
|
38
|
OSR1 and SPAK cooperatively modulate Sertoli cell support of mouse spermatogenesis. Sci Rep 2016; 6:37205. [PMID: 27853306 PMCID: PMC5112561 DOI: 10.1038/srep37205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
We investigated the role of oxidative stress-responsive kinase-1 (OSR1) and STE20 (sterile 20)/SPS1-related proline/alanine-rich kinase (SPAK), upstream regulators of the Na+-K+-2Cl− cotransporter (NKCC1)—essential for spermatogenesis—in mouse models of male fertility. Global OSR1+/− gene mutations, but not global SPAK−/− or Sertoli cell (SC)-specific OSR1 gene knockout (SC-OSR1−/−), cause subfertility with impaired sperm function and are associated with reduced abundance of phosphorylated (p)-NKCC1 but increased p-SPAK expression in testicular tissue and spermatozoa. To dissect further in a SC-specific manner the compensatory effect of OSR1 and SPAK in male fertility, we generated SC-OSR1−/− and SPAK−/− double knockout (DKO) male mice. These are infertile with defective spermatogenesis, presenting a SC-only-like syndrome. Disrupted meiotic progression and increased germ cell apoptosis occurred in the first wave of spermatogenesis. The abundance of total and p-NKCC1 was significantly decreased in the testicular tissues of DKO mice. These results indicate that OSR1 and SPAK cooperatively regulate NKCC1-dependent spermatogenesis in a SC-restricted manner.
Collapse
|
39
|
Chen Q, Deng T, Han D. Testicular immunoregulation and spermatogenesis. Semin Cell Dev Biol 2016; 59:157-165. [DOI: 10.1016/j.semcdb.2016.01.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 12/22/2022]
|
40
|
Bulldan A, Dietze R, Shihan M, Scheiner-Bobis G. Non-classical testosterone signaling mediated through ZIP9 stimulates claudin expression and tight junction formation in Sertoli cells. Cell Signal 2016; 28:1075-85. [PMID: 27164415 DOI: 10.1016/j.cellsig.2016.04.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 11/27/2022]
Abstract
In the classical signaling pathway, testosterone regulates gene expression by activating the cytosolic/nuclear androgen receptor. In the non-classical pathway, testosterone activates cytosolic signaling cascades that are normally triggered by growth factors. The nature of the receptor involved in this signaling pathway is a source of controversy. In the Sertoli cell line 93RS2, which lacks the classical AR, we determined that testosterone stimulates the non-classical signaling pathway, characterized by the phosphorylation of Erk1/2 and transcription factors CREB and ATF-1. We also demonstrated that testosterone increases the expression of the tight junction (TJ) proteins claudin-1 and claudin-5. Both of these proteins are known to be essential constituents of TJs between Sertoli cells, and as a consequence of their increased expression transepithelial resistance across Sertoli cell monolayers is increased. ZIP9 is a Zn(2+)transporter that was recently shown to be a membrane-bound testosterone receptor. Silencing its expression in 93RS2 Sertoli cells by siRNA completely prevents Erk1/2, CREB, and ATF-1 phosphorylation as well the stimulation of claudin-1 and -5 expression and TJ formation between neighboring cells. The study presented here demonstrates for the first time that in Sertoli cells testosterone acts through the receptor ZIP9 to trigger the non-classical signaling cascade, resulting in increased claudin expression and TJ formation. Since TJ formation is a prerequisite for the maintenance of the blood-testis barrier, the testosterone/ZIP9 effects might be significant for male physiology. Further assessment of these interactions will help to supplement our knowledge concerning the mechanism by which testosterone plays a role in male fertility.
Collapse
Affiliation(s)
- Ahmed Bulldan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Raimund Dietze
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Mazen Shihan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany.
| |
Collapse
|
41
|
França LR, Hess RA, Dufour JM, Hofmann MC, Griswold MD. The Sertoli cell: one hundred fifty years of beauty and plasticity. Andrology 2016; 4:189-212. [PMID: 26846984 DOI: 10.1111/andr.12165] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/30/2015] [Accepted: 01/04/2016] [Indexed: 12/18/2022]
Abstract
It has been one and a half centuries since Enrico Sertoli published the seminal discovery of the testicular 'nurse cell', not only a key cell in the testis, but indeed one of the most amazing cells in the vertebrate body. In this review, we begin by examining the three phases of morphological research that have occurred in the study of Sertoli cells, because microscopic anatomy was essentially the only scientific discipline available for about the first 75 years after the discovery. Biochemistry and molecular biology then changed all of biological sciences, including our understanding of the functions of Sertoli cells. Immunology and stem cell biology were not even topics of science in 1865, but they have now become major issues in our appreciation of Sertoli cell's role in spermatogenesis. We end with the universal importance and plasticity of function by comparing Sertoli cells in fish, amphibians, and mammals. In these various classes of vertebrates, Sertoli cells have quite different modes of proliferation and epithelial maintenance, cystic vs. tubular formation, yet accomplish essentially the same function but in strikingly different ways.
Collapse
Affiliation(s)
- L R França
- Laboratory of Cellular Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,National Institute for Amazonian Research (INPA), Manaus, Amazonas, Brazil
| | - R A Hess
- Reproductive Biology and Toxicology, Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA
| | - J M Dufour
- Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - M C Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M D Griswold
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
42
|
Hong F, Wang Y, Zhou Y, Zhang Q, Ge Y, Chen M, Hong J, Wang L. Exposure to TiO2 Nanoparticles Induces Immunological Dysfunction in Mouse Testitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:346-55. [PMID: 26720763 DOI: 10.1021/acs.jafc.5b05262] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Although TiO2 nanoparticles (NPs) as endocrine disruptors have been demonstrated to be able to cross the blood-testis barriers and induce reproductive toxicity in male animals, whether the reproductive toxicity of male animals due to exposure to endocrine disruptor TiO2 NPs is related to immunological dysfunction in the testis remains not well understood. This study determined whether the reproductive toxicity and immunological dysfunction induced by exposure to TiO2 NPs is associated with activation or inhibition of TAM/TLR-mediated signal pathway in mouse testis. The results showed that male mice exhibited significant reduction of fertility, infiltration of inflammatory cells, rarefaction, apoptosis, and/or necrosis of spermatogenic cells and Sertoli cells due to TiO2 NPs. Furthermore, these were associated with decreased expression of Tyro3 (-18.16 to -66.6%), Axl (-14.7 to -57.99%), Mer (-7.98 to -72.62%), and IκB (-11.25 to -63.16%), suppression of cytokine signaling (SOCS) 1 (-21.99 to -73.8%) and SOCS3 (-8.11 to -34.86%), and increased expression of Toll-like receptor (TLR)-3 (21.4-156.03%), TLR-4 (37.0-109.87%), nuclear factor-κB (14.75-69.34%), interleukin (IL)-lβ (46.15-123.08%), IL-6 (2.54-81.98%), tumor necrosis factor-α (6.95-88.39%), interferon (IFN)-α (2.54-37.25%), and IFN-β (10.19-80.56%), which are involved in the immune environment in the testis. The findings showed that reproductive toxicity of male mice induced by exposure to endocrine disruptor TiO2 NPs may be associated with biomarkers of impairment of immune environment or dysfunction of TAM/TLR3-mediated signal pathway in mouse testitis. Therefore, the potential risks to reproductive health should be attended, especially in those who are occupationally exposed to TiO2 NPs.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Yajing Wang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Yingjun Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Qi Zhang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Yushuang Ge
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Ming Chen
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University , Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University , Huaian 223300, China
- School of Life Sciences, Huaiyin Normal University , Huaian 223300, China
| | - Jie Hong
- Medical College of Soochow University , Suzhou 215123, China
| | - Ling Wang
- Library of Soochow University , Suzhou 215123, China
| |
Collapse
|
43
|
Lambeth LS, Morris KR, Wise TG, Cummins DM, O'Neil TE, Cao Y, Sinclair AH, Doran TJ, Smith CA. Transgenic Chickens Overexpressing Aromatase Have High Estrogen Levels but Maintain a Predominantly Male Phenotype. Endocrinology 2016; 157:83-90. [PMID: 26556534 DOI: 10.1210/en.2015-1697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogens play a key role in sexual differentiation of both the gonads and external traits in birds. The production of estrogen occurs via a well-characterized steroidogenic pathway, which is a multistep process involving several enzymes, including cytochrome P450 aromatase. In chicken embryos, the aromatase gene (CYP19A1) is expressed female-specifically from the time of gonadal sex differentiation. Ectopic overexpression of aromatase in male chicken embryos induces gonadal sex reversal, and male embryos treated with estradiol become feminized; however, this is not permanent. To test whether a continuous supply of estrogen in adult chickens could induce stable male to female sex reversal, 2 transgenic male chickens overexpressing aromatase were generated using the Tol2/transposase system. These birds had robust ectopic aromatase expression, which resulted in the production of high serum levels of estradiol. Transgenic males had female-like wattle and comb growth and feathering, but they retained male weights, displayed leg spurs, and developed testes. Despite the small sample size, this data strongly suggests that high levels of circulating estrogen are insufficient to maintain a female gonadal phenotype in adult birds. Previous observations of gynandromorph birds and embryos with mixed sex chimeric gonads have highlighted the role of cell autonomous sex identity in chickens. This might imply that in the study described here, direct genetic effects of the male chromosomes largely prevailed over the hormonal profile of the aromatase transgenic birds. This data therefore support the emerging view of at least partial cell autonomous sex development in birds. However, a larger study will confirm this intriguing observation.
Collapse
Affiliation(s)
- Luke S Lambeth
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Kirsten R Morris
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Terry G Wise
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - David M Cummins
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Terri E O'Neil
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Yu Cao
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Andrew H Sinclair
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Timothy J Doran
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Craig A Smith
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
44
|
Mruk DD, Cheng CY. The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev 2015; 36:564-91. [PMID: 26357922 PMCID: PMC4591527 DOI: 10.1210/er.2014-1101] [Citation(s) in RCA: 442] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
Spermatogenesis is the cellular process by which spermatogonia develop into mature spermatids within seminiferous tubules, the functional unit of the mammalian testis, under the structural and nutritional support of Sertoli cells and the precise regulation of endocrine factors. As germ cells develop, they traverse the seminiferous epithelium, a process that involves restructuring of Sertoli-germ cell junctions, as well as Sertoli-Sertoli cell junctions at the blood-testis barrier. The blood-testis barrier, one of the tightest tissue barriers in the mammalian body, divides the seminiferous epithelium into 2 compartments, basal and adluminal. The blood-testis barrier is different from most other tissue barriers in that it is not only comprised of tight junctions. Instead, tight junctions coexist and cofunction with ectoplasmic specializations, desmosomes, and gap junctions to create a unique microenvironment for the completion of meiosis and the subsequent development of spermatids into spermatozoa via spermiogenesis. Studies from the past decade or so have identified the key structural, scaffolding, and signaling proteins of the blood-testis barrier. More recent studies have defined the regulatory mechanisms that underlie blood-testis barrier function. We review here the biology and regulation of the mammalian blood-testis barrier and highlight research areas that should be expanded in future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, New York, New York 10065
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, New York, New York 10065
| |
Collapse
|
45
|
Testosterone deficiency induced by progressive stages of diabetes mellitus impairs glucose metabolism and favors glycogenesis in mature rat Sertoli cells. Int J Biochem Cell Biol 2015; 66:1-10. [DOI: 10.1016/j.biocel.2015.07.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 06/10/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022]
|
46
|
Fijak M, Damm LJ, Wenzel JP, Aslani F, Walecki M, Wahle E, Eisel F, Bhushan S, Hackstein H, Baal N, Schuler G, Konrad L, Rafiq A, O'Hara L, Smith LB, Meinhardt A. Influence of Testosterone on Inflammatory Response in Testicular Cells and Expression of Transcription Factor Foxp3 in T Cells. Am J Reprod Immunol 2015; 74:12-25. [DOI: 10.1111/aji.12363] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/02/2015] [Indexed: 01/31/2023] Open
Affiliation(s)
- Monika Fijak
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Lara-Jil Damm
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Jan-Per Wenzel
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Ferial Aslani
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Magdalena Walecki
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Eva Wahle
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Florian Eisel
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine; Justus-Liebig-University; Giessen Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine; Justus-Liebig-University; Giessen Germany
| | - Gerhard Schuler
- Clinic for Obstetrics; Gynecology and Andrology of Large and Small Animals; Faculty of Veterinary Medicine; Justus-Liebig-University; Giessen Germany
| | - Lutz Konrad
- Department of Obstetrics and Gynaecology; Faculty of Medicine; Justus-Liebig-University; Giessen Germany
| | - Amir Rafiq
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| | - Laura O'Hara
- MRC Centre for Reproductive Health; University of Edinburgh; Edinburgh UK
| | - Lee B. Smith
- MRC Centre for Reproductive Health; University of Edinburgh; Edinburgh UK
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology; Justus-Liebig-University; Giessen Germany
| |
Collapse
|
47
|
Mohapatra S, Chakraborty T, Miyagawa S, Zhou L, Ohta K, Iguchi T, Nagahama Y. Steroid responsive regulation of IFNγ2 alternative splicing and its possible role in germ cell proliferation in medaka. Mol Cell Endocrinol 2015; 400:61-70. [PMID: 25458697 DOI: 10.1016/j.mce.2014.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 11/23/2022]
Abstract
Interferon gamma (IFNγ) is an active player in estrogen dependent immuno-regulation of fish. The present work was aimed to characterize the alternatively spliced isoforms of IFNγ2 in the gonadal sex development in medaka. Phylogenetic analysis demonstrated that IFNγ2a and 2b were clustered with fish specific interferon gamma. Our in vitro promoter and mini-genome analysis data confirmed that alternative splicing of IFNγ2 is regulated by estrogens and androgens. Tissue distribution, quantitative PCR and ISH data demonstrated ubiquitous expression of IFNγ2a, while IFNγ2b was only expressed predominantly in female germ cells than males. This was further confirmed by germ cell specific GFP signals in the IFNγ2b-GFP over-expressed embryos and specific induction of IFNγ2b expression in the BrdU positive cells. All together our data suggest that steroid responsive alternatively spliced IFNγ2b isoforms might have some indirect roles in germ cell proliferation and thus can be an important candidate for immuno-reproductive interaction studies.
Collapse
Affiliation(s)
- Sipra Mohapatra
- South Ehime Fisheries Research Center, Ehime University, 798-4206, Matsuyama, Japan; Key Laboratory of Freshwater Fish Reproduction and Development, Southwest University, Chongqing, 400715, China; Division of Molecular Environmental Endocrinology, National Institute of Natural Sciences, Okazaki, 444-8585, Japan
| | - Tapas Chakraborty
- South Ehime Fisheries Research Center, Ehime University, 798-4206, Matsuyama, Japan; Division of Molecular Environmental Endocrinology, National Institute of Natural Sciences, Okazaki, 444-8585, Japan.
| | - Shinichi Miyagawa
- Division of Molecular Environmental Endocrinology, National Institute of Natural Sciences, Okazaki, 444-8585, Japan
| | - Linyan Zhou
- Key Laboratory of Freshwater Fish Reproduction and Development, Southwest University, Chongqing, 400715, China
| | - Kohei Ohta
- South Ehime Fisheries Research Center, Ehime University, 798-4206, Matsuyama, Japan
| | - Taisen Iguchi
- Division of Molecular Environmental Endocrinology, National Institute of Natural Sciences, Okazaki, 444-8585, Japan
| | - Yoshitaka Nagahama
- South Ehime Fisheries Research Center, Ehime University, 798-4206, Matsuyama, Japan.
| |
Collapse
|
48
|
|
49
|
Redgrove KA, McLaughlin EA. The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword. Front Immunol 2014; 5:534. [PMID: 25386180 PMCID: PMC4209867 DOI: 10.3389/fimmu.2014.00534] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (CT) is the most prevalent bacterial sexually transmitted infection in the world, with more than 100 million cases reported annually. While there have been extensive studies into the adverse effects that CT infection has on the female genital tract, and on the subsequent ability of these women to conceive, studies into the consequences on male fertility have been limited and controversial. This is in part due to the asymptomatic nature of the infection, where it is estimated that 50% of men with Chlamydia fail to show any symptoms. It is accepted, however, that acute and/or persistent CT infection is the causative agent for conditions such as urethritis, epididymitis, epididymo-orchitis, and potentially prostatitis. As with most infections, the immune system plays a fundamental role in the body’s attempts to eradicate the infection. The first and most important immune response to Chlamydia infection is a local one, whereby immune cells such as leukocytes are recruited to the site of infections, and subsequently secrete pro-inflammatory cytokines and chemokines such as interferon gamma. Immune cells also work to initiate and potentiate chronic inflammation through the production of reactive oxygen species (ROS), and the release of molecules with degradative properties including defensins, elastase, collagenase, cathespins, and lysozyme. This long-term inflammation can lead to cell proliferation (a possible precursor to cancer), tissue remodeling, and scarring, as well as being linked to the onset of autoimmune responses in genetically disposed individuals. This review will focus on the ability of the immune system to recognize and clear acute and persistent chlamydial infections in the male genital tract, and on the paradoxical damage that chronic inflammation resulting from the infection can cause on the reproductive health of the individual.
Collapse
Affiliation(s)
- Kate A Redgrove
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| | - Eileen A McLaughlin
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| |
Collapse
|
50
|
Walker WH. Testosterone signaling and the regulation of spermatogenesis. SPERMATOGENESIS 2014; 1:116-120. [PMID: 22319659 DOI: 10.4161/spmg.1.2.16956] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 05/31/2011] [Indexed: 12/12/2022]
Abstract
Spermatogenesis and male fertility are dependent upon the presence of testosterone in the testis. In the absence of testosterone or the androgen receptor, spermatogenesis does not proceed beyond the meiosis stage. The major cellular target and translator of testosterone signals to developing germ cells is the Sertoli cell. In the Sertoli cell, testosterone signals can be translated directly to changes in gene expression (the classical pathway) or testosterone can activate kinases that may regulate processes required to maintain spermatogenesis (the non-classical pathway). Contributions of the classical and non-classical testosterone signaling pathways to the maintenance of spermatogenesis are discussed. Studies that may further elaborate the mechanisms by with the pathways support spermatogenesis are proposed.
Collapse
Affiliation(s)
- William H Walker
- Center for Research in Reproductive Physiology; Department of Obstetrics, Gynecology and Reproduction Sciences; Magee Women's Research Institute; University of Pittsburgh; Pittsburgh, PA USA
| |
Collapse
|