1
|
Chen Y, Liu B, Tao S, Liu L, Gao J, Liang Y, Dong W, Zhou D. CITED2 Binding to EP300 Regulates Human Spermatogonial Stem Cell Proliferation and Survival Through HSPA6. Stem Cells Int 2025; 2025:2362489. [PMID: 40313859 PMCID: PMC12045681 DOI: 10.1155/sci/2362489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/03/2025] Open
Abstract
Spermatogonial stem cells (SSCs) are essential for the initiation and continuation of spermatogenesis, a process fundamental to male fertility. Despite extensive studies on mouse SSCs, the mechanisms governing self-renewal and differentiation in human SSCs remain to be elucidated. This study investigated the regulatory mechanisms of SSCs by analyzing single-cell sequencing data from the GEO dataset of human testis. Analysis revealed dominant expression of CITED2 in human SSCs. Reduction of CITED2 levels in hSSC lines significantly inhibited proliferation and increased apoptosis. Protein interaction prediction and immunoprecipitation identified interactions between CITED2 and EP300 in SSC lines. RNA sequencing results indicated that CITED2 knockdown significantly affected the MAPK pathway and the HSPA6 gene. Overexpression of HSPA6 mitigated the proliferative and apoptotic changes provoked by CITED2 downregulation. These findings provide novel insights into the regulatory and functional mechanisms of CITED2-mediated hSSC development.
Collapse
Affiliation(s)
- Yongzhe Chen
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Bang Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Sisi Tao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Lvjun Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Jianxin Gao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Ying Liang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Weilei Dong
- Gynecology and Obstetrics and Reproductive Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- MOE Key Lab of Rare Pediatric Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Research Department, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| |
Collapse
|
2
|
Niedenberger BA, Belcher HA, Gilbert EA, Thomas MA, Geyer CB. Utilization of the QuPath open-source software platform for analysis of mammalian spermatogenesis†. Biol Reprod 2025; 112:583-599. [PMID: 39817641 PMCID: PMC11911557 DOI: 10.1093/biolre/ioaf011] [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: 09/11/2024] [Revised: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 01/18/2025] Open
Abstract
The adult mammalian testis is filled with seminiferous tubules, which contain somatic Sertoli cells along with germ cells undergoing all phases of spermatogenesis. During spermatogenesis in postnatal mice, male germ cells undergo at least 17 different nomenclature changes as they proceed through mitosis as spermatogonia (=8), meiosis as spermatocytes (=6), and spermiogenesis as spermatids (=3). Adding to this complexity, combinations of germ cells at each of these stages of development are clumped together along the length of the seminiferous tubules. Due to this, considerable expertise is required for investigators to accurately analyze changes in spermatogenesis in animals that have spontaneous mutations, have been genetically modified (transgenic or knockout/knockin), or have been treated with pharmacologic agents. Here, we leverage our laboratory's expertise in spermatogenesis to optimize the open-source "Quantitative Pathology & Bioimage Analysis" software platform for automated analyses of germ and somatic cell populations in both the developing and adult mammalian testis.
Collapse
Affiliation(s)
- Bryan A Niedenberger
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Heather A Belcher
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Emma A Gilbert
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Matthew A Thomas
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| |
Collapse
|
3
|
Li J, Shen L, Wang K, Wu S, Wang Y, Pan Y, Chen S, Zhao T, Zhao Y, Niu L, Chen L, Zhang S, Zhu L, Gan M. Biogenesis of stress granules and their role in the regulation of stress-induced male reproduction disorders. Cell Commun Signal 2025; 23:84. [PMID: 39948590 PMCID: PMC11827146 DOI: 10.1186/s12964-025-02054-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Stress granules (SGs) are conserved messenger ribonucleoprotein (mRNP) granules that form through rapid coalescence in the cytoplasm of eukaryotic cells under stressful environments. These dynamic membrane-free organelles can respond to a variety of both intracellular and extracellular stressors. Studies have shown that stress conditions such as heat stress, arsenite exposure, and hypoxic stress can induce SGs formation. The formation of SGs helps mitigates the effects of environmental stimuli on cells, protects them from damage, and promotes cell survival. This paper focuses on the biogenesis of SGs and summarizes the role in regulating environmental stress-induced male reproductive disorders, with the aim of exploring SGs as a potential means of mitigating male reproduction disorders. Numerous studies have demonstrated that the detrimental effects of environmental stress on germ cells can be effectively suppressed by regulating the formation and timely disassembly of SGs. Therefore, regulating the phosphorylation of eIF2α and the assembly and disassembly of SGs could offer a promising therapeutic strategy to alleviate the impacts of environmental stress on male reproduction health.
Collapse
Affiliation(s)
- Jiaxin Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Kai Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shuang Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuheng Pan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Siyu Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ting Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
4
|
Delgouffe E, Silva SM, Chalmel F, Cools W, Raets C, Tilleman K, T'Sjoen G, Baert Y, Goossens E. Partial rejuvenation of the spermatogonial stem cell niche after gender-affirming hormone therapy in trans women. eLife 2025; 13:RP94825. [PMID: 39773877 PMCID: PMC11706602 DOI: 10.7554/elife.94825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Although the impact of gender-affirming hormone therapy (GAHT) on spermatogenesis in trans women has already been studied, data on its precise effects on the testicular environment is poor. Therefore, this study aimed to characterize, through histological and transcriptomic analysis, the spermatogonial stem cell niche of 106 trans women who underwent standardized GAHT, comprising estrogens and cyproterone acetate. A partial dedifferentiation of Sertoli cells was observed, marked by the co-expression of androgen receptor and anti-Müllerian hormone which mirrors the situation in peripubertal boys. The Leydig cells also exhibited a distribution analogous to peripubertal tissue, accompanied by a reduced insulin-like factor 3 expression. Although most peritubular myoid cells expressed alpha-smooth muscle actin 2, the expression pattern was disturbed. Besides this, fibrosis was particularly evident in the tubular wall and the lumen was collapsing in most participants. A spermatogenic arrest was also observed in all participants. The transcriptomic profile of transgender tissue confirmed a loss of mature characteristics - a partial rejuvenation - of the spermatogonial stem cell niche and, in addition, detected inflammation processes occurring in the samples. The present study shows that GAHT changes the spermatogonial stem cell niche by partially rejuvenating the somatic cells and inducing fibrotic processes. These findings are important to further understand how estrogens and testosterone suppression affect the testis environment, and in the case of orchidectomized testes as medical waste material, their potential use in research.
Collapse
Affiliation(s)
- Emily Delgouffe
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| | - Samuel Madureira Silva
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| | - Frédéric Chalmel
- Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (IRSET), Université de RennesRennesFrance
| | - Wilfried Cools
- Core facility, Support for Quantitative and Qualitative Research (SQUARE), Vrije Universiteit BrusselBrusselsBelgium
| | - Camille Raets
- Core facility, Support for Quantitative and Qualitative Research (SQUARE), Vrije Universiteit BrusselBrusselsBelgium
| | - Kelly Tilleman
- Department for Reproductive Medicine, Ghent University HospitalGhentBelgium
| | - Guy T'Sjoen
- Department of Endocrinology and Center for Sexology and Gender, Ghent University HospitalGhentBelgium
| | - Yoni Baert
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
- In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit BrusselBrusselsBelgium
| | - Ellen Goossens
- Biology of the Testis (BITE) Laboratory, Genetics, Reproduction and Development (GRAD) Research Group, Vrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
5
|
Bu T, Wang L, Wu X, Gao S, Yun D, Mao B, Li L, Sun F, Cheng CY. Interacting Fat1 and Dchs Planar Cell Polarity Proteins Supported by Fjx1 Serve as Heterodimeric Intercellular Bridges Crucial to Support Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:355-374. [PMID: 40301264 DOI: 10.1007/978-3-031-82990-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Studies of the planar cell polarity (PCP) protein complexes Fat1/Fjx1 and Dchs/Fjx1 that form heterotypic interacting bridges of Fat1-Dchs between adjacent cells to confer PCP, as noted in Drosophila, are also found in mammalian cells and tissues as orthologs, such as in Sertoli cells and condensed spermatids in the seminiferous epithelium of the testis. Recent studies have shown that these two interacting PCP protein complexes are also crucial regulators of microtubule and actin dynamics, modulating the polymerization of both microtubules and actin filaments in the testis. In this review, we provide a brief update and thought-provoking concept on the PCP core proteins and the associated downstream signaling pathways utilized by PCP proteins to confer PCP and regulation of the microtubule and actin cytoskeletons in the testis. However, we focus on recent data in the field on the Fat1/Fjx1 and Dchs/Fjx1 protein complexes, which are also heterotypic interacting protein complexes, and their functional role in modulating the microtubule and actin cytoskeletal organization. Based on these recent findings, we formulate a hypothetic model depicting the role of these two PCP protein complexes in modulating the timely "opening" and "closing" of the blood-testis barrier (BTB) formed by adjacent Sertoli cells near the base of the seminiferous epithelium. Additionally, these two PCP protein complexes also modulate cytoskeletal dynamics between Sertoli cells and condensed spermatids to support haploid spermatid transport across the seminiferous epithelium during their structural transformation through spermiogenesis, and their eventual release at spermiation during the epithelial cycle of spermatogenesis. This hypothetical model will provide a useful framework for designing functional experiments to understand the role of PCP proteins in supporting spermatogenesis.
Collapse
Affiliation(s)
- Tiao Bu
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang City, Guangdong, China
| | - Lingling Wang
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Gao
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Damin Yun
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baiping Mao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run-run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Brezin F, Busiah K, Leroy C, Fiot E, Bensignor C, Amouroux C, Caquard M, Cartault A, Castets S, Delcour C, Devernay M, Feigerlova E, Hoarau M, Lebon-Labich B, Lambert AS, Rouleau S, Trouvin MA, Vautier V, Martinerie L. Endocrine management of transgender adolescents: Expert consensus of the french society of pediatric endocrinology and diabetology working group. Arch Pediatr 2024:S0929-693X(24)00176-3. [PMID: 39551654 DOI: 10.1016/j.arcped.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/25/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Requests for hormonal transition in minors are increasing. To date, there is no national recommendation to guide these practices in France. Therefore, the SFEDP (French Society of Pediatric Endocrinology and Diabetology) has commissioned a group of experts to draft the first national consensus on this topic. METHOD Each chapter was prepared by one to three authors who conducted a literature review, and it was then reviewed and revised by the group as many times as necessary to achieve a consensus position. The final document was reviewed by a group of external experts. RESULTS A consensus position was reached regarding the multi-professional nature of support for trans youth, the prescription of molecules aimed at inhibiting endogenous hormone secretion, and the use of gender-affirming hormone therapies, as well as the importance of offering gamete preservation. Non-hormonal aspects of support and various considerations, including ethical ones, were also discussed. CONCLUSION This work constitutes an initial set of recommendations for professionals involved in the hormonal transition of trans youth. Additional recommendations under the auspices of the French High Authority for Health would be worthy of being drafted, involving all relevant stakeholders to establish comprehensive official national guidelines that would secure the support and rights of these young individuals, especially those under 16 years old, as well as the professionals involved in their care.
Collapse
Affiliation(s)
- François Brezin
- Unité D'endocrinologie et Diabétologie Pédiatrique, Centre Hospitalier Régional Universitaire, Service de Pédiatrie 1, Strasbourg, France.
| | - Kanetee Busiah
- Unité D'endocrinologie, Diabétologie et Obésité Pédiatrique, Département Femmes-Mères-Enfants, Centre Hospitalier Universitaire, Lausanne, Suisse
| | - Clara Leroy
- Unité D'endocrinologie et Diabétologie Pédiatrique, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire, Lille, France
| | - Elodie Fiot
- Service d'Endocrinologie et Diabétologie Pédiatrique, EPPAT, Hôpital Universitaire Robert-Debré, GHU APHP Nord, Paris 75019, France
| | - Candace Bensignor
- Service de pédiatrie, Centre Hospitalier Universitaire, Dijon, France
| | - Cyril Amouroux
- Unité de Néphrologie et Endocrinologie, Service de Pédiatrie Multidisplinaire, Centre Hospitalier Universitaire, Montpellier, France; Département de Pédagogie, Faculté de Médecine, Université de Montpellier, Montpellier, France
| | - Marylène Caquard
- Service de Pédiatrie, Centre Hospitalier Universitaire, Nantes, France
| | - Audrey Cartault
- Unité D'endocrinologie, Génétique, Maladies Osseuses et Gynécologie Pédiatrique, Hôpital d'enfants, Centre Hospitalier Universitaire, Toulouse, France
| | - Sarah Castets
- Service de Pédiatrie Multidisplinaire, Hôpital Timone, Assistance Publique - Hôpitaux de Marseille, Marseille, France
| | - Clémence Delcour
- Service de Gynécologie, EPPAT, Hôpital Universitaire Robert-Debré, GHU APHP Nord, Paris 75019, France; Faculté de Santé, UFR de Médecine, Paris Cité, Paris, France
| | - Marie Devernay
- Unité de Médecine Pour Adolescents, Hôpital Armand Trousseau, APHP, Paris, France
| | - Eva Feigerlova
- Service D'endocrinologie, Diabète et Nutrition, Centre Hospitalier Universitaire, Nancy-Brabois, France
| | - Marie Hoarau
- Service de Pédiatrie Générale, Centre Hospitalier Universitaire, Saint Pierre, La Réunion, France
| | - Béatrice Lebon-Labich
- Service de Médecine Infantile, Hôpital d'enfant, Centre Hospitalier Régional Universitaire, Nancy-Brabois, France
| | - Anne-Sophie Lambert
- Service D'endocrinologie et Diabète de L'enfant, Hôpital Kremlin-Bicêtre, APHP, Paris, France
| | - Stéphanie Rouleau
- Unité D'endocrinologie, Gynécologie et Diabétologie Pédiatrique, Service de Pédiatrie, Centre Hospitalier Universitaire, Angers, France
| | - Marie-Agathe Trouvin
- Service D'endocrinologie et Diabète de L'enfant, Hôpital Kremlin-Bicêtre, APHP, Paris, France
| | - Vanessa Vautier
- Unité de Diabétologie, Endocrinologie et Gynécologie Pédiatrique, Hôpital des Enfants, Centre Hospitalier Universitaire, Bordeaux, France
| | - Laetitia Martinerie
- Service d'Endocrinologie et Diabétologie Pédiatrique, EPPAT, Hôpital Universitaire Robert-Debré, GHU APHP Nord, Paris 75019, France; Faculté de Santé, UFR de Médecine, Paris Cité, Paris, France.
| |
Collapse
|
7
|
Mao BP, Pan M, Shan Y, Wang YN, Li H, Wu J, Zhu X, Hu E, Cheng CY, Shangguan W. Katanin regulatory subunit B1 (KATNB1) regulates BTB dynamics through changes in cytoskeletal organization. FASEB J 2024; 38:e70049. [PMID: 39275889 DOI: 10.1096/fj.202400966r] [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: 04/27/2024] [Revised: 07/31/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024]
Abstract
In this study, we have explored the role of the KATNB1 gene, a microtubule-severing protein, in the seminiferous epithelium of the rat testis. Our data have shown that KATNB1 expressed in rat brain, testes, and Sertoli cells. KATNB1 was found to co-localize with α-tubulin showing a unique stage-specific distribution across the seminiferous epithelium. Knockdown of KATNB1 by RNAi led to significant disruption of the tight junction (TJ) permeability barrier function in primary Sertoli cells cultured in vitro with an established functional TJ-barrier, as well as perturbations in the microtubule and actin cytoskeleton organization. The disruption in these cytoskeletal structures, in turn, led to improper distribution of TJ and basal ES proteins essential for maintaining the Sertoli TJ function. More importantly, overexpression of KATNB1 in the testis in vivo was found to block cadmium-induced blood-testis barrier (BTB) disruption and testis injury. KATNB1 exerted its promoting effects on BTB and spermatogenesis through corrective spatiotemporal expression of actin- and microtubule-based regulatory proteins by maintaining the proper organization of cytoskeletons in the testis, illustrating its plausible therapeutic implication. In summary, Katanin regulatory subunit B1 (KATNB1) plays a crucial role in BTB and spermatogenesis through its effects on the actin- and microtubule-based cytoskeletons in Sertoli cells and testis, providing important insights into male reproductive biology.
Collapse
Affiliation(s)
- Bai-Ping Mao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Mingdong Pan
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Shan
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Nan Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huitao Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinhan Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuanjing Zhu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ende Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangning Shangguan
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Huang Z, Li N, Ji X, Zhou D, Wang R, Zhao X, Wang S, Zhang H, Huang C, Lin G. EEF1B2 regulates the proliferation and apoptosis of human spermatogonial stem cell lines through TAF4B. Heliyon 2024; 10:e36467. [PMID: 39281470 PMCID: PMC11401128 DOI: 10.1016/j.heliyon.2024.e36467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Background Spermatogonial stem cells (SSCs) are essential for male fertility, maintaining sperm production throughout life. While mouse SSCs have been studied extensively, the mechanisms regulating human SSCs are less understood. Objectives To investigate the role of EEF1B2 in regulating human SSC proliferation and apoptosis. Material and methods Single cell RNA sequencing (scRNA-seq) analysis was utilized to investigate the differentially expressed genes of SSC. The distribution of EEF1B2 in the human testis was examined using immunofluorescence and immunohistochemistry techniques. Cell proliferation, DNA replication, and self-renewal were analyzed using CCK8, EdU, Western blot, and flow cytometry. RNA sequencing was employed to analyze the downstream target molecules and signaling pathways of EEF1B2. Results In this study, we analyzed single-cell sequencing data from human testicular samples and identified EEF1B2 as a protein highly expressed in SSCs, with expression decreasing during development. Immunohistochemistry and immunofluorescence confirmed this pattern and showed co-localization with the proliferation marker KI67. Knockdown of EEF1B2 in human SSC lines impaired proliferation and viability, reducing self-renewal proteins like PLZF and CCNE1. RNA sequencing revealed decreased TAF4B following EEF1B2 knockdown, which could be rescued by replenishing TAF4B. Testicular SSCs from non-obstructive azoospermia (NOA) patients also showed reduced EEF1B2. Discussion and conclusion Our findings reveal a novel regulatory mechanism involving EEF1B2 and TAF4B in human SSCs, suggesting EEF1B2 deficiency may contribute to male infertility.
Collapse
Affiliation(s)
- Zenghui Huang
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Ning Li
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China, Hunan, 410008, China
| | - Xiren Ji
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan, 410000, China
| | - Ruijun Wang
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Xingguo Zhao
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Shuangyao Wang
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
| | - Huan Zhang
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Chuan Huang
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| | - Ge Lin
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, China
| |
Collapse
|
9
|
Sanchez NDRR, Ritagliati C, Kopf GS, Kretschmer S, Buck J, Levin LR. The uniqueness of on-demand male contraception. Mol Aspects Med 2024; 97:101281. [PMID: 38805792 PMCID: PMC11167369 DOI: 10.1016/j.mam.2024.101281] [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: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Because nearly half of pregnancies worldwide are unintended, available contraceptive methods are inadequate. Moreover, due to the striking imbalance between contraceptive options available for men compared to the myriad of options available to women, there is an urgent need for new methods of contraception for men. This review summarizes ongoing efforts to develop male contraceptives highlighting the unique aspects particular to on-demand male contraception, where a man takes a contraceptive only when and as often as needed.
Collapse
Affiliation(s)
| | - Carla Ritagliati
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Dai P, Ding M, Yu J, Gao Y, Wang M, Ling J, Dong S, Zhang X, Zeng X, Sun X. The Male Reproductive Toxicity Caused by 2-Naphthylamine Was Related to Testicular Immunity Disorders. TOXICS 2024; 12:342. [PMID: 38787121 PMCID: PMC11126000 DOI: 10.3390/toxics12050342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
2-naphthylamine (NAP) was classified as a group I carcinogen associated with bladder cancer. The daily exposure is mostly from cigarette and E-cigarette smoke. NAP can lead to testicular atrophy and interstitial tissue hyperplasia; however, the outcomes of NAP treatment on spermatogenesis and the associated mechanisms have not been reported. The study aimed to investigate the effect of NAP on spermatogenesis and sperm physiologic functions after being persistently exposed to NAP at 5, 20, and 40 mg/kg for 35 days. We found that sperm motility, progressive motility, sperm average path velocity, and straight-line velocity declined remarkably in the NAP (40 mg/kg) treated group, and the sperm deformation rate rose upon NAP administration. The testis immunity- and lipid metabolism-associated processes were enriched from RNA-sequence profiling. Plvap, Ccr7, Foxn1, Trim29, Sirpb1c, Cfd, and Lpar4 involved in testis immunity and Pnliprp1 that inhibit triglyceride and cholesterol absorption were confirmed to rise dramatically in the NAP-exposed group. The increased total cholesterol and CD68 levels were observed in the testis from the NAP-exposed group. Gpx5, serving as an antioxidant in sperm plasma, and Semg1, which contributes to sperm progressive motility, were both down-regulated. We concluded that the short-term exposure to NAP caused reproductive toxicity, primarily due to the inflammatory abnormality in the testis.
Collapse
Affiliation(s)
- Pengyuan Dai
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Mengqian Ding
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Jingyan Yu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Yuan Gao
- Experimental Animal Center, Nantong University, Nantong 226001, China;
| | - Miaomiao Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Jie Ling
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Shijue Dong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China; (P.D.); (M.D.); (J.Y.); (M.W.); (J.L.); (S.D.); (X.Z.)
| | - Xiaoli Sun
- Center for Reproductive Medicine, The Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| |
Collapse
|
11
|
Wang Z, Wang Y, Zhou T, Chen S, Morris D, Magalhães RDM, Li M, Wang S, Wang H, Xie Y, McSwiggin H, Oliver D, Yuan S, Zheng H, Mohammed J, Lai EC, McCarrey JR, Yan W. The rapidly evolving X-linked MIR-506 family fine-tunes spermatogenesis to enhance sperm competition. eLife 2024; 13:RP90203. [PMID: 38639482 PMCID: PMC11031087 DOI: 10.7554/elife.90203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Despite rapid evolution across eutherian mammals, the X-linked MIR-506 family miRNAs are located in a region flanked by two highly conserved protein-coding genes (SLITRK2 and FMR1) on the X chromosome. Intriguingly, these miRNAs are predominantly expressed in the testis, suggesting a potential role in spermatogenesis and male fertility. Here, we report that the X-linked MIR-506 family miRNAs were derived from the MER91C DNA transposons. Selective inactivation of individual miRNAs or clusters caused no discernible defects, but simultaneous ablation of five clusters containing 19 members of the MIR-506 family led to reduced male fertility in mice. Despite normal sperm counts, motility, and morphology, the KO sperm were less competitive than wild-type sperm when subjected to a polyandrous mating scheme. Transcriptomic and bioinformatic analyses revealed that these X-linked MIR-506 family miRNAs, in addition to targeting a set of conserved genes, have more targets that are critical for spermatogenesis and embryonic development during evolution. Our data suggest that the MIR-506 family miRNAs function to enhance sperm competitiveness and reproductive fitness of the male by finetuning gene expression during spermatogenesis.
Collapse
Affiliation(s)
- Zhuqing Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Yue Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Sheng Chen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Dayton Morris
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | | | - Musheng Li
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Shawn Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Hetan Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Hayden McSwiggin
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Jaaved Mohammed
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - John R McCarrey
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San AntonioSan AntonioUnited States
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
- Department of Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
12
|
Zhang G, Zhi W, Ye F, Xiong D, Zhang Y, Liu F, Zhao Y, Du X, Wu Y, Hou M, Liu J, Wei J, Silang Y, Xu W, Zeng J, Chen S, Liu W. Systematic analyses of the factors influencing sperm quality in patients with SARS-CoV-2 infection. Sci Rep 2024; 14:8132. [PMID: 38584153 PMCID: PMC10999436 DOI: 10.1038/s41598-024-58797-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/03/2024] [Indexed: 04/09/2024] Open
Abstract
To figure out how does SARS-CoV-2 affect sperm parameters and what influencing factors affect the recovery of sperm quality after infection? We conducted a prospective cohort study and initially included 122 men with SARS-CoV-2 infection. The longest time to track semen quality after infection is 112 days and 58 eligible patients were included in our study eventually. We subsequently exploited a linear mixed-effects model to statistically analyze their semen parameters at different time points before and after SARS-CoV-2 infection. Semen parameters were significantly reduced after SARS-CoV-2 infection, including total sperm count (211 [147; 347] to 167 [65.0; 258], P < 0.001), sperm concentration (69.0 [38.8; 97.0] to 51.0 [25.5; 71.5], P < 0.001), total sperm motility (57.5 [52.3; 65.0] to 51.0 [38.5; 56.8], P < 0.001), progressive motility (50.0 [46.2; 58.0] to 45.0 [31.5; 52.8], P < 0.001). The parameters displayed the greatest diminution within 30 days after SARS-CoV-2 infection, gradually recovered thereafter, and exhibited no significant difference after 90 days compared with prior to COVID-19 infection. In addition, the patients in the group with a low-grade fever showed a declining tendency in semen parameters, but not to a significant degree, whereas those men with a moderate or high fever produced a significant drop in the same parameters. Semen parameters were significantly reduced after SARS-CoV-2 infection, and fever severity during SARS-CoV-2 infection may constitute the main influencing factor in reducing semen parameters in patients after recovery, but the effect is reversible and the semen parameters gradually return to normal with the realization of a new spermatogenic cycle.
Collapse
Affiliation(s)
- Guohui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, China
| | - Weiwei Zhi
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Fei Ye
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Dongsheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Yanan Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Fulin Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, China
| | - Yuhong Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610500, China
| | - Xinrong Du
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yang Wu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Mingxia Hou
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Jiu Liu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Jiajing Wei
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Yangzhong Silang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China
| | - Wenming Xu
- Department of Obstetrics and Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiuzhi Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China.
| | - Shiqi Chen
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China.
| | - Weixin Liu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, China.
| |
Collapse
|
13
|
Wang L, Yan M, Bu T, Wu X, Li L, Silvestrini B, Sun F, Cheng CY, Chen H. Map-1a regulates Sertoli cell BTB dynamics through the cytoskeletal organization of microtubule and F-actin. Reprod Biol Endocrinol 2024; 22:36. [PMID: 38570783 PMCID: PMC10988971 DOI: 10.1186/s12958-024-01204-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Microtubule-associated protein 1a (Map1a) is a microtubule (MT) regulatory protein that binds to the MT protofilaments in mammalian cells to promote MT stabilization. Maps work with MT cleavage proteins and other MT catastrophe-inducing proteins to confer MT dynamics to support changes in the Sertoli cell shape to sustain spermatogenesis. However, no functional studies are found in the literature to probe its role in spermatogenesis. Using an RNAi approach, coupled with the use of toxicant-induced testis (in vivo)- and Sertoli cell (in vitro)-injury models, RNA-Seq analysis, transcriptome profiling, and relevant bioinformatics analysis, immunofluorescence analysis, and pertinent biochemical assays for cytoskeletal organization, we have delineated the functional role of Map1a in Sertoli cells and testes. Map1a was shown to support MT structural organization, and its knockdown (KD) also perturbed the structural organization of actin, vimentin, and septin cytoskeletons as these cytoskeletons are intimately related, working in concert to support spermatogenesis. More importantly, cadmium-induced Sertoli cell injury that perturbed the MT structural organization across the cell cytoplasm was associated with disruptive changes in the distribution of Map1a and a surge in p-p38-MAPK (phosphorylated p38-mitogen-activated protein kinase) expression but not total p38-MAPK. These findings thus support the notion that p-p38-MAPK activation is involved in cadmium-induced Sertoli cell injury. This conclusion was supported by studies using doramapimod, a specific p38-MAPK phosphorylation (activation) inhibitor, which was capable of restoring the cadmium-induced disruptive structural organization of MTs across the Sertoli cell cytoplasm. In summary: this study provides mechanistic insights regarding restoration of toxicant-induced Sertoli cell and testis injury and male infertility.
Collapse
Affiliation(s)
- Lingling Wang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Ming Yan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Tiao Bu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang 325027, Wenzhou, China
| | - Bruno Silvestrini
- Faculty of Pharmacy, University of Rome La Sapienza, P. Le Aldo Moro 5, 00185, Rome, Italy
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| | - C Yan Cheng
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Hao Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
14
|
Wang Z, Wang Y, Zhou T, Chen S, Morris D, Magalhães RDM, Li M, Wang S, Wang H, Xie Y, McSwiggin H, Oliver D, Yuan S, Zheng H, Mohammed J, Lai EC, McCarrey JR, Yan W. The Rapidly Evolving X-linked miR-506 Family Finetunes Spermatogenesis to Enhance Sperm Competition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544876. [PMID: 37398484 PMCID: PMC10312769 DOI: 10.1101/2023.06.14.544876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Despite rapid evolution across eutherian mammals, the X-linked miR-506 family miRNAs are located in a region flanked by two highly conserved protein-coding genes (Slitrk2 and Fmr1) on the X chromosome. Intriguingly, these miRNAs are predominantly expressed in the testis, suggesting a potential role in spermatogenesis and male fertility. Here, we report that the X-linked miR-506 family miRNAs were derived from the MER91C DNA transposons. Selective inactivation of individual miRNAs or clusters caused no discernable defects, but simultaneous ablation of five clusters containing nineteen members of the miR-506 family led to reduced male fertility in mice. Despite normal sperm counts, motility and morphology, the KO sperm were less competitive than wild-type sperm when subjected to a polyandrous mating scheme. Transcriptomic and bioinformatic analyses revealed that these X-linked miR-506 family miRNAs, in addition to targeting a set of conserved genes, have more targets that are critical for spermatogenesis and embryonic development during evolution. Our data suggest that the miR-506 family miRNAs function to enhance sperm competitiveness and reproductive fitness of the male by finetuning gene expression during spermatogenesis.
Collapse
Affiliation(s)
- Zhuqing Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Yue Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Sheng Chen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Dayton Morris
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | | | - Musheng Li
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Shawn Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Hetan Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Hayden McSwiggin
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jaaved Mohammed
- Department of Developmental Biology, Memorial Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA
| | - Eric C. Lai
- Department of Developmental Biology, Memorial Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA
| | - John R. McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Li L, Feng T, Wu R, Zhang Y, Wang N, Wu M, Pang Y, Yang S, Yang A, Zhang D, Hao G, Zhang R. The role of total antioxidant capacity and malondialdehyde of seminal plasma in the association between air pollution and sperm quality. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122324. [PMID: 37544399 DOI: 10.1016/j.envpol.2023.122324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/08/2023]
Abstract
Accumulating evidence has suggested that men exposed to air pollution are associated with decreased sperm quality, and seminal plasma plays a pivotal role in maintaining sperm viability. However, the role of seminal plasma in air pollution related sperm quality decline remain unestablished. In current study, we recruited 524 participants from couples who underwent in vitro fertilization treatment due to female factors at a fertility clinic in China from March to August 2020. Conventional sperm parameters, total antioxidant capacity (T-AOC), malondialdehyde (MDA) and testosterone were measured using semen samples. The six main air pollutants (PM2.5, PM10, NO2, SO2, CO, O3) during four key periods of sperm development (meiotic stage, spermiogenesis stage, epididymal stage and total sperm cycle period) were estimated using inverse distance weighting method. Multiple linear regression models were employed to investigate the exposure-outcome relationships. And we found that PM10 exposures were negatively related to sperm total motility and the exposures of PM2.5 and PM10 were inversely associated with sperm progressive motility during epididymal stage. Furthermore, PM2.5 and PM10 exposures were positively associated with seminal plasma MDA and PM10 was negatively related to seminal plasma T-AOC during epididymal stage. PM2.5, PM10 and CO exposures during total sperm cycle period might relate to increased seminal plasma testosterone. Mediation analysis indicated seminal plasma MDA and T-AOC partially mediated PM10 associated reduction of sperm motility during epididymal stage. Our study suggested MDA and T-AOC of seminal plasma played a role in air pollution associated decline of sperm motility.
Collapse
Affiliation(s)
- Lipeng Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China; Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Tengfei Feng
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ruiting Wu
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China
| | - Yaling Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China
| | - Ning Wang
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China
| | - Sujuan Yang
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Aimin Yang
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dengsuo Zhang
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Guimin Hao
- Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China; Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, 050017, Hebei, PR China.
| |
Collapse
|
16
|
Giannattasio T, Testa E, Faieta M, Lampitto M, Nardozi D, di Cecca S, Russo A, Barchi M. The proper interplay between the expression of Spo11 splice isoforms and the structure of the pseudoautosomal region promotes XY chromosomes recombination. Cell Mol Life Sci 2023; 80:279. [PMID: 37682311 PMCID: PMC10491539 DOI: 10.1007/s00018-023-04912-7] [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: 04/15/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023]
Abstract
XY chromosome missegregation is relatively common in humans and can lead to sterility or the generation of aneuploid spermatozoa. A leading cause of XY missegregation in mammals is the lack of formation of double-strand breaks (DSBs) in the pseudoautosomal region (PAR), a defect that may occur in mice due to faulty expression of Spo11 splice isoforms. Using a knock-in (ki) mouse that expresses only the single Spo11β splice isoform, here we demonstrate that by varying the genetic background of mice, the length of chromatin loops extending from the PAR axis and the XY recombination proficiency varies. In spermatocytes of C57Spo11βki/- mice, in which loops are relatively short, recombination/synapsis between XY is fairly normal. In contrast, in cells of C57/129Spo11βki/- males where PAR loops are relatively long, formation of DSBs in the PAR (more frequently the Y-PAR) and XY synapsis fails at a high rate, and mice produce sperm with sex-chromosomal aneuploidy. However, if the entire set of Spo11 splicing isoforms is expressed by a wild type allele in the C57/129 background, XY recombination and synapsis is recovered. By generating a Spo11αki mouse model, we prove that concomitant expression of SPO11β and SPO11α isoforms, boosts DSB formation in the PAR. Based on these findings, we propose that SPO11 splice isoforms cooperate functionally in promoting recombination in the PAR, constraining XY asynapsis defects that may arise due to differences in the conformation of the PAR between mouse strains.
Collapse
Affiliation(s)
- Teresa Giannattasio
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Erika Testa
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Monica Faieta
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Matteo Lampitto
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Daniela Nardozi
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Stefano di Cecca
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy
| | - Antonella Russo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marco Barchi
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, Rome, Italy.
- Department of Biomedical Science, Lady of Good Counsel University, Tirana, Albania.
| |
Collapse
|
17
|
Kirsanov O, Johnson TA, Niedenberger BA, Malachowski TN, Hale BJ, Chen Q, Lackford B, Wang J, Singh A, Schindler K, Hermann BP, Hu G, Geyer CB. Retinoic acid is dispensable for meiotic initiation but required for spermiogenesis in the mammalian testis. Development 2023; 150:dev201638. [PMID: 37350382 PMCID: PMC10357014 DOI: 10.1242/dev.201638] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
Retinoic acid (RA) is the proposed mammalian 'meiosis inducing substance'. However, evidence for this role comes from studies in the fetal ovary, where germ cell differentiation and meiotic initiation are temporally inseparable. In the postnatal testis, these events are separated by more than 1 week. Exploiting this difference, we discovered that, although RA is required for spermatogonial differentiation, it is dispensable for the subsequent initiation, progression and completion of meiosis. Indeed, in the absence of RA, the meiotic transcriptome program in both differentiating spermatogonia and spermatocytes entering meiosis was largely unaffected. Instead, transcripts encoding factors required during spermiogenesis were aberrant during preleptonema, and the subsequent spermatid morphogenesis program was disrupted such that no sperm were produced. Taken together, these data reveal a RA-independent model for male meiotic initiation.
Collapse
Affiliation(s)
- Oleksandr Kirsanov
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Taylor A. Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Bryan A. Niedenberger
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Taylor N. Malachowski
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Benjamin J. Hale
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Qing Chen
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Brad Lackford
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Jiajia Wang
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Anukriti Singh
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Guang Hu
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Christopher B. Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
18
|
Zhao X, Huang Z, Chen Y, Zhou Q, Zhu F, Zhang H, Zhou D. MAGEB2-Mediated Degradation of EGR1 Regulates the Proliferation and Apoptosis of Human Spermatogonial Stem Cell Lines. Stem Cells Int 2023; 2023:3610466. [PMID: 37304127 PMCID: PMC10256451 DOI: 10.1155/2023/3610466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Spermatogonial stem cells are committed to initiating and maintaining male spermatogenesis, which is the foundation of male fertility. Understanding the mechanisms underlying SSC fate decisions is critical for controlling spermatogenesis and male fertility. However, the key molecules and mechanisms responsible for regulating human SSC development are not clearly understood. Here, we analyzed normal human testis single-cell sequencing data from the GEO dataset (GSE149512 and GSE112013). Melanoma antigen gene B2 (MAGEB2) was found to be predominantly expressed in human SSCs and further validated by immunohistology. Overexpression of MAGEB2 in SSC lines severely weakened cell proliferation and promoted apoptosis. Further, using protein interaction prediction, molecular docking, and immunoprecipitation, we found that MAGEB2 interacted with early growth response protein 1 (EGR1) in SSC lines. Reexpression of EGR1 in MAGEB2 overexpression cells partially rescued decreased cell proliferation. Furthermore, MAGEB2 was shown to be downregulated in specific NOA patients, implying that abnormal expression of MAGEB2 may impair spermatogenesis and male fertility. Our results offer new insights into the functional and regulatory mechanisms in MAGEB2-mediated human SSC line proliferation and apoptosis.
Collapse
Affiliation(s)
- Xueheng Zhao
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Zenghui Huang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Yongzhe Chen
- First Affiliated Hospital of University of South China, Hengyang, Hunan 421000, China
| | - Qianyin Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Fang Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Huan Zhang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Dai Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
- College of Life Sciences, Hunan Normal University, Changsha, Hunan 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan 410000, China
| |
Collapse
|
19
|
Johnson TA, Niedenberger BA, Kirsanov O, Harrington EV, Malachowski T, Geyer CB. Differential responsiveness of spermatogonia to retinoic acid dictates precocious differentiation but not meiotic entry during steady-state spermatogenesis†. Biol Reprod 2023; 108:822-836. [PMID: 36708226 PMCID: PMC10183363 DOI: 10.1093/biolre/ioad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 01/29/2023] Open
Abstract
The foundation of mammalian spermatogenesis is provided by undifferentiated spermatogonia, which comprise of spermatogonial stem cells (SSCs) and transit-amplifying progenitors that differentiate in response to retinoic acid (RA) and are committed to enter meiosis. Our laboratory recently reported that the foundational populations of SSCs, undifferentiated progenitors, and differentiating spermatogonia are formed in the neonatal testis in part based on their differential responsiveness to RA. Here, we expand on those findings to define the extent to which RA responsiveness during steady-state spermatogenesis in the adult testis regulates the spermatogonial fate. Our results reveal that both progenitor and differentiating spermatogonia throughout the testis are capable of responding to exogenous RA, but their resulting fates were quite distinct-undifferentiated progenitors precociously differentiated and proceeded into meiosis on a normal timeline, while differentiating spermatogonia were unable to hasten their entry into meiosis. This reveals that the spermatogonia responding to RA must still complete the 8.6 day differentiation program prior to their entry into meiosis. Addition of exogenous RA enriched testes with preleptotene and pachytene spermatocytes one and two seminiferous cycles later, respectively, supporting recent clinical studies reporting increased sperm production and enhanced fertility in subfertile men on long-term RA analog treatment. Collectively, our results reveal that a well-buffered system exists within mammalian testes to regulate spermatogonial RA exposure, that exposed undifferentiated progenitors can precociously differentiate, but must complete a normal-length differentiation program prior to entering meiosis, and that daily RA treatments increased the numbers of advanced germ cells by directing undifferentiated progenitors to continuously differentiate.
Collapse
Affiliation(s)
- Taylor A Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Bryan A Niedenberger
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Oleksandr Kirsanov
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Ellen V Harrington
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Taylor Malachowski
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NCUSA
| |
Collapse
|
20
|
Chua SC, Yovich SJ, Hinchliffe PM, Yovich JL. Male Clinical Parameters (Age, Stature, Weight, Body Mass Index, Smoking History, Alcohol Consumption) Bear Minimal Relationship to the Level of Sperm DNA Fragmentation. J Pers Med 2023; 13:jpm13050759. [PMID: 37240929 DOI: 10.3390/jpm13050759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
This retrospective cohort study reports on 1291 males who were the partners of women presenting with infertility requiring assisted reproduction and who had sperm DNA fragmentation (SDF) levels measured by the Halosperm test. These men provided clinical and biometric details which included their age, stature, weight, and body mass index (BMI). Of these men, 562 (43.5%) provided detailed historical records of their smoking and alcohol histories. The aim of this study was to determine whether any clinical and biometric parameters, or main lifestyle factors, had any influence on SDF. We found that the only clinical parameter with a direct correlation was that of advancing age (r = 0.064, p = 0.02), but none of the biometric parameters of stature, weight, or BMI showed any significant correlation. In respect to lifestyle, there were significant correlations with smoking history, but not in the way we expected. Our data showed significantly elevated SDF levels among non-smokers (p = 0.03) compared with smokers. We also found that, among the non-smokers, ex-smokers had higher SDF levels (p = 0.03). With respect to alcohol, consumers did not show any significant differences in SDF levels. These lifestyle findings did not show any significant relevance with respect to an SDF level of <15% or ≥15%. Furthermore, logistic regression analysis excluded age as a confounder in these lifestyle findings. It is therefore concluded that, apart from age, both clinical and lifestyle aspects have minimal relevance to SDF.
Collapse
Affiliation(s)
- Shiao Chuan Chua
- PIVET Medical Centre, Perth, WA 6007, Australia
- Hospital Shah Alam, Shah Alam 40000, Selangor, Malaysia
| | | | | | - John Lui Yovich
- PIVET Medical Centre, Perth, WA 6007, Australia
- Faculty of Health Sciences, School of Medicine, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
21
|
Gobé C, Ialy-Radio C, Pierre R, Cocquet J. Generation and Characterization of a Transgenic Mouse That Specifically Expresses the Cre Recombinase in Spermatids. Genes (Basel) 2023; 14:genes14050983. [PMID: 37239343 DOI: 10.3390/genes14050983] [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: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Spermiogenesis is the step during which post-meiotic cells, called spermatids, undergo numerous morphological changes and differentiate into spermatozoa. Thousands of genes have been described to be expressed at this stage and could contribute to spermatid differentiation. Genetically-engineered mouse models using Cre/LoxP or CrispR/Cas9 are the favored approaches to characterize gene function and better understand the genetic basis of male infertility. In the present study, we produced a new spermatid-specific Cre transgenic mouse line, in which the improved iCre recombinase is expressed under the control of the acrosomal vesicle protein 1 gene promoter (Acrv1-iCre). We show that Cre protein expression is restricted to the testis and only detected in round spermatids of stage V to VIII seminiferous tubules. The Acrv1-iCre line can conditionally knockout a gene during spermiogenesis with a > 95% efficiency. Therefore, it could be useful to unravel the function of genes during the late stage of spermatogenesis, but it can also be used to produce an embryo with a paternally deleted allele without causing early spermatogenesis defects.
Collapse
Affiliation(s)
- Clara Gobé
- Université Paris Cité, INSERM, CNRS, Institut Cochin, F-75014 Paris, France
| | - Côme Ialy-Radio
- Université Paris Cité, INSERM, CNRS, Institut Cochin, F-75014 Paris, France
| | - Rémi Pierre
- Université Paris Cité, INSERM, CNRS, Institut Cochin, F-75014 Paris, France
- Homologous Recombination, Embryo Transfer and Cryopreservation Facility, Cochin Institute, University of Paris, F-75006 Paris, France
| | - Julie Cocquet
- Université Paris Cité, INSERM, CNRS, Institut Cochin, F-75014 Paris, France
| |
Collapse
|
22
|
The Impact of COVID-19 Vaccines on Male Semen Parameters: A Retrospective Cohort Study. Andrologia 2023. [DOI: 10.1155/2023/7826568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
The emergence of SARS-CoV-2 and the subsequent COVID-19 pandemic necessitated the development of adequate vaccines. Despite vaccines being demonstrated to be safe and effective for preventing severe disease and death, vaccine hesitancy remains. Reasons include concerns over adverse effects on male fertility, which have not been widely investigated. Therefore, this study is aimed at determining the impact of COVID-19 vaccination on semen parameters in a retrospective cohort study of South African males undergoing fertility assessment. The patients for this study were adult men who have previously undergone routine semen analysis for fertility assessment at Androcryos Andrology Laboratory (Johannesburg, South Africa) between March 2021 and March 2022. They also received vaccination within 3 months following a semen analysis and underwent a second semen analysis any time post-COVID-19 vaccination. From 277 records analysed, 46 patients met the inclusion criteria, receiving the Pfizer-BioNTech (BNT162b1) (63%), Johnson and Johnson (JNJ-78436735/Ad26.COV2S) (34.8%), and the AstraZeneca (AZD1222) (2.2%) vaccines. Sperm concentration significantly increased postvaccination (
), with no significant changes in semen pH, volume, total sperm count, progressive motility, normal sperm morphology, or chromatin condensation. Results were not influenced by age, type of vaccine received, and the number of days following vaccination, as depicted by multiple regression analysis. In conclusion, there is no evidence of a negative impact of COVID-19 vaccination on male semen parameters, which is consistent with the emerging literature on COVID-19 vaccination and male fertility. COVID-19 vaccinations should not be dismissed based on fear of adverse effects on male fertility parameters.
Collapse
|
23
|
Li N, Zhou Q, Yi Z, Zhang H, Zhou D. Ubiquitin protein E3 ligase ASB9 suppresses proliferation and promotes apoptosis in human spermatogonial stem cell line by inducing HIF1AN degradation. Biol Res 2023; 56:4. [PMID: 36683111 PMCID: PMC9869568 DOI: 10.1186/s40659-023-00413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Spermatogonial stem cells (SSCs) are critical for sustaining spermatogenesis. Even though several regulators of SSC have been identified in rodents, the regulatory mechanism of SSC in humans has yet to be discovered. METHODS To explore the regulatory mechanisms of human SSCs, we analyzed publicly available human testicular single-cell sequencing data and found that Ankyrin repeat and SOCS box protein 9 (ASB9) is highly expressed in SSCs. We examined the expression localization of ASB9 using immunohistochemistry and overexpressed ASB9 in human SSC lines to explore its role in SSC proliferation and apoptosis. Meanwhile, we used immunoprecipitation to find the target protein of ASB9 and verified its functions. In addition, we examined the changes in the distribution of ASB9 in non-obstructive azoospermia (NOA) patients using Western blot and immunofluorescence. RESULTS The results of uniform manifold approximation and projection (UMAP) clustering and pseudotime analysis showed that ASB9 was highly expressed in SSCs, and its expression gradually increased during development. The immunohistochemical and dual-color immunofluorescence results displayed that ASB9 was mainly expressed in nonproliferating SSCs. Overexpression of ASB9 in the SSC line revealed significant inhibition of cell proliferation and increased apoptosis. We predicted the target proteins of ASB9 and verified that hypoxia-inducible factor 1-alpha inhibitor (HIF1AN), but not creatine kinase B-type (CKB), has a direct interaction with ASB9 in human SSC line using protein immunoprecipitation experiments. Subsequently, we re-expressed HIF1AN in ASB9 overexpressing cells and found that HIF1AN reversed the proliferative and apoptotic changes induced by ASB9 overexpression. In addition, we found that ABS9 was significantly downregulated in some NOA patients, implying a correlation between ASB9 dysregulation and impaired spermatogenesis. CONCLUSION ASB9 is predominantly expressed in human SSCs, it affects the proliferation and apoptotic process of the SSC line through HIF1AN, and its abnormal expression may be associated with NOA.
Collapse
Affiliation(s)
- Ning Li
- grid.216417.70000 0001 0379 7164Operating Department of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164Xiangya Nursing School, Central South University, Changsha, 410013 Hunan China
| | - Qianyin Zhou
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Zhang Yi
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Huan Zhang
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Dai Zhou
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China ,grid.411427.50000 0001 0089 3695College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan China ,Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, 410013 Hunan China
| |
Collapse
|
24
|
Meikar O, Majoral D, Heikkinen O, Valkama E, Leskinen S, Rebane A, Ruusuvuori P, Toppari J, Mäkelä JA, Kotaja N. STAGETOOL, a Novel Automated Approach for Mouse Testis Histological Analysis. Endocrinology 2022; 164:6868503. [PMID: 36461763 PMCID: PMC9780747 DOI: 10.1210/endocr/bqac202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Spermatogenesis is a complex differentiation process that takes place in the seminiferous tubules. A specific organization of spermatogenic cells within the seminiferous epithelium enables a synchronous progress of germ cells at certain steps of differentiation on the spermatogenic pathway. This can be observed in testis cross-sections where seminiferous tubules can be classified into distinct stages of constant cellular composition (12 stages in the mouse). For a detailed analysis of spermatogenesis, these stages have to be individually observed from testis cross-sections. However, the recognition of stages requires special training and expertise. Furthermore, the manual scoring is laborious considering the high number of tubule cross-sections that have to be analyzed. To facilitate the analysis of spermatogenesis, we have developed a convolutional deep neural network-based approach named "STAGETOOL." STAGETOOL analyses histological images of 4',6-diamidine-2'-phenylindole dihydrochloride (DAPI)-stained mouse testis cross-sections at ×400 magnification, and very accurately classifies tubule cross-sections into 5 stage classes and cells into 9 categories. STAGETOOL classification accuracy for stage classes of seminiferous tubules of a whole-testis cross-section is 99.1%. For cellular level analysis the F1 score for 9 seminiferous epithelial cell types ranges from 0.80 to 0.98. Furthermore, we show that STAGETOOL can be applied for the analysis of knockout mouse models with spermatogenic defects, as well as for automated profiling of protein expression patterns. STAGETOOL is the first fluorescent labeling-based automated method for mouse testis histological analysis that enables both stage and cell-type recognition. While STAGETOOL qualitatively parallels an experienced human histologist, it outperforms humans time-wise, therefore representing a major advancement in male reproductive biology research.
Collapse
Affiliation(s)
| | | | - Olli Heikkinen
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, 20520 Turku, Finland
| | - Eero Valkama
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, 20520 Turku, Finland
| | - Sini Leskinen
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, 20520 Turku, Finland
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Pekka Ruusuvuori
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, 20520 Turku, Finland
- Department of Pediatrics, Turku University Hospital, 20520 Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, 20520 Turku, Finland
| | - Juho-Antti Mäkelä
- Correspondence: Juho-Antti Mäkelä, PhD, Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland. ; or Noora Kotaja, PhD, Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Noora Kotaja
- Correspondence: Juho-Antti Mäkelä, PhD, Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland. ; or Noora Kotaja, PhD, Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| |
Collapse
|
25
|
Wu X, Yun D, Sang M, Liu J, Zhou L, Shi J, Wang L, Bu T, Li L, Huang Y, Lin D, Sun F, Cheng CY. Defects of microtubule cytoskeletal organization in NOA human testes. Reprod Biol Endocrinol 2022; 20:154. [PMID: 36329464 PMCID: PMC9632130 DOI: 10.1186/s12958-022-01026-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022] Open
Abstract
The importance of actin and microtubule (MT) cytoskeletons in testis function in rodents is known to some extent, but its role in the etiology of azoospermia in humans remains unexplored. Here, we examined if MT cytoskeleton was defective in NOA (non-obstructive azoospermia) testes versus normal human testes based on histopathological, immunofluorescence (IF), and scRNA-Seq transcriptome profiling. Testis biopsy samples from n = 6 normal men versus n = 3 Sertoli cell only (SCO) and n = 3 MA (meiotic arrest) of NOA patients were used for histopathological analysis. IF analysis was also used to examine MT organization across the seminiferous epithelium, investigating the likely involvement of microtubule-associated proteins (MAPs). scRNA-Seq transcriptome profiling datasets from testes of 3 SCO patients versus 3 normal men in public domain in Gene Expression Omnibus (GEO) Sample (GSM) with identifiers were analyzed to examine relevant genes that regulate MT dynamics. NOA testes of MA and SCO patients displayed notable defects in MT organization across the epithelium with extensive truncation, mis-alignments and appeared as collapsed structures near the base of the tubules. These changes are in contrast to MTs in testes of normal men. scRNA-Seq analyses revealed considerable loss of spermatogenesis capacity in SCO testes of NOA patients versus normal men. An array of genes that support MT dynamics displayed considerable changes in expression and in spatial distribution. In summary, defects in MT cytoskeleton were noted in testes of NOA (SCO) patients, possibly mediated by defective spatial expression and/or distribution of MAPs. These changes, in turn, may impede spermatogenesis in SCO testes of NOA patients.
Collapse
Affiliation(s)
- Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Damin Yun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Mengmeng Sang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jianpeng Liu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Tiao Bu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Linxi Li
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - YingYing Huang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Dengfeng Lin
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, 226001, Jiangsu, China.
- Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, 1230 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
26
|
H4K5 Butyrylation Coexist with Acetylation during Human Spermiogenesis and Are Retained in the Mature Sperm Chromatin. Int J Mol Sci 2022; 23:ijms232012398. [PMID: 36293256 PMCID: PMC9604518 DOI: 10.3390/ijms232012398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Male germ cells experience a drastic chromatin remodeling through the nucleo-histone to nucleo-protamine (NH-NP) transition necessary for proper sperm functionality. Post-translational modifications (PTMs) of H4 Lys5, such as acetylation (H4K5ac), play a crucial role in epigenetic control of nucleosome disassembly facilitating protamine incorporation into paternal DNA. It has been shown that butyrylation on the same residue (H4K5bu) participates in temporal regulation of NH-NP transition in mice, delaying the bromodomain testis specific protein (BRDT)-dependent nucleosome disassembly and potentially marking retained nucleosomes. However, no information was available so far on this modification in human sperm. Here, we report a dual behavior of H4K5bu and H4K5ac in human normal spermatogenesis, suggesting a specific role of H4K5bu during spermatid elongation, coexisting with H4K5ac although with different starting points. This pattern is stable under different testicular pathologies, suggesting a highly conserved function of these modifications. Despite a drastic decrease of both PTMs in condensed spermatids, they are retained in ejaculated sperm, with 30% of non-colocalizing nucleosome clusters, which could reflect differential paternal genome retention. Whereas no apparent effect of these PTMs was observed associated with sperm quality, their presence in mature sperm could entail a potential role in the zygote.
Collapse
|
27
|
Xu J, He K, Zhou Y, Zhao L, Lin Y, Huang Z, Xie N, Yue J, Tang Y. The effect of SSRIs on Semen quality: A systematic review and meta-analysis. Front Pharmacol 2022; 13:911489. [PMID: 36188547 PMCID: PMC9519136 DOI: 10.3389/fphar.2022.911489] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are widely used for a variety of diseases, and their impact on semen quality is unclear. We performed a systematic search in PubMed and Embase, and after a strict screening, we included 4 studies with a total of 222 male participants. In result, SSRIs reduced normal sperm morphology (95% CI [−16.29, −3.77], p = 0.002), sperm concentration (95%CI [−43.88, −4.18], p = 0.02), sperm motility (95%CI [−23.46, −0.47], p = 0.04) and sperm DNA fragmentation index (DFI) (95% CI [6.66,21.93], p = 0.0002), without a statistically significant effect on semen volume (95%CI [−0.75,0.65], p = 0.89). Moreover, the impact on both sperm morphology and sperm concentration were observed within the 3-month period of SSRIs use. In general, our meta-analysis showed that SSRIs have a negative effect on semen quality. More larger, randomized, well-controlled clinical studies should be conducted to support our conclusion.
Collapse
Affiliation(s)
- Jiarong Xu
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Kancheng He
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yihong Zhou
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
| | - Liangyu Zhao
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
| | - Yuteng Lin
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
| | - Zihao Huang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
| | - Nengqing Xie
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
| | - Jihui Yue
- Department of Psychiatry, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- *Correspondence: Jihui Yue, ; Yuxin Tang,
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- *Correspondence: Jihui Yue, ; Yuxin Tang,
| |
Collapse
|
28
|
Zhang T, He H, Wei Y, Geng W, Zhai J. Vitamin C supplementation rescued meiotic arrest of spermatocytes in Balb/c mice exposed to BDE-209. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113846. [PMID: 35853364 DOI: 10.1016/j.ecoenv.2022.113846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 06/15/2023]
Abstract
Deca-brominated diphenyl ether (BDE-209) is a ubiquitous industrial chemical as brominated flame retardant (BFRs). Exposure to BDE-209 has been clearly associated with male reproductive disorders. However, the meiotic arrest mechanism of spermatocytes exposed to BDE-209 is still unclear. The present work aimed to explore the protective effect of vitamin C on BDE-209-induced meiotic arrest of spermatocytes and its possible mechanism. Vitamin C (100 mg/kg BW) was administered to BDE-209-exposed (80 mg/kg BW) male Balb/c mice once daily by intraperitoneal injection for 2 weeks. Our results showed that vitamin C played male reproductive protection effects as showed by attenuated BDE-209-induced testicular damage, and reduced sperm abnormality rate. Vitamin C also attenuated BDE-209-induced increase in SOD and MDA in testes and GC-2 spd cells. Moreover, vitamin C promoted meiotic prophase in BDE-209-induced mice, with suppressed γ-H2AX, restored DMC1, RAD51, and crossover marker MLH1 levels, and prevented BDE-209-induced DNA impairment. In addition, vitamin C supplementation also interfered with BDE-209-induced upregulation of testicular H3K4me3 through inhibition of KDM5s capacity and decreasing ferrous ion concentration. Furthermore, ferrous sulfate pretreatment could partially restore the expression of H3K4me3 via maintaining the concentration of ferrous ions. Taken together, vitamin C exerts a potential therapeutic agent for preventing BDE-209-induced reproductive toxicity with meiotic arrest, which is attributed to its antioxidant and electron donor properties, as well as, modulation of ferrous ion levels and demethylation of H3K4me3.
Collapse
Affiliation(s)
- Taifa Zhang
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei 230032, China
| | - Huan He
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei 230032, China
| | - Yu Wei
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei 230032, China
| | - Wenfeng Geng
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei 230032, China
| | - Jinxia Zhai
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Meishan Rd 81, Hefei 230032, China.
| |
Collapse
|
29
|
Belladelli F, Chen T, Basran S, Greenberg DR, Del Giudice F, Mulloy E, Chen CH, Cheng YS, Salonia A, Eisenberg ML. The Association between Monthly, Yearly, and Lifetime Cannabis Use, and Semen Parameters in Asian-American Men. World J Mens Health 2022:40.e61. [PMID: 36047080 DOI: 10.5534/wjmh.220106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Medicinal and recreational cannabis use has grown exponentially, however, its effect on testicular function and spermatogenesis remains uncertain. The aim of this study was to evaluate the association between cannabis use and semen parameters in a cohort of Asian-American men with unknown fertility. MATERIALS AND METHODS Asian men were recruited to complete an online survey and submit a semen sample. Semen analysis, demographic data, lifestyle factors, and cannabis use habits were collected. Linear and logistic regression analyses were used to determine. RESULTS Among the 112 men included in this study, 51 used cannabis at least once in their lifetime, 30 men used cannabis at least once in the last 12 months, and 26 men used cannabis at least once in the last 30 days. Adjusted linear regression analyses identified an association between cannabis use in the previous 30 days and worse sperm morphology (β: -0.45, p=0.025) and sperm motility (β: -1.64, p=0.016). However, when stratifying by subfertile semen quality (i.e., WHO criteria), no association was identified between semen quality and cannabis use. Lower sperm morphology and motility are partially associated with recent cannabis use, while all other semen parameters are not. CONCLUSIONS We did not observe any consistent associations between cannabis use on any semen parameters in Asian-American men. Further studies within the field are needed to explore racial and ethnic differences in semen quality and lifestyle factors.
Collapse
Affiliation(s)
- Federico Belladelli
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy.,Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Tony Chen
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Satvir Basran
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Daniel R Greenberg
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Francesco Del Giudice
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Urology, University Sapienza, Rome, Italy
| | - Evan Mulloy
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yu-Sheng Cheng
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology; URI; IRCCS Ospedale San Raffaele, Milan, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Michael L Eisenberg
- Department of Urology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
30
|
Verratti V, Mrakic-Sposta S, Fusi J, Sabovic I, Franzoni F, Pietrangelo T, Bondi D, Dall’Acqua S, Daniele S, Scarfò G, Di Giulio C, Garolla A. Fertility Impairment after Trekking at High Altitude: A Proof of Mechanisms on Redox and Metabolic Seminal Changes. Int J Mol Sci 2022; 23:9066. [PMID: 36012330 PMCID: PMC9409093 DOI: 10.3390/ijms23169066] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Many authors described negative but reversible effects of high-altitude hypoxic exposure on animal and human fertility in terms of sperm concentration, function, and biochemical alterations. The aim of this study was to evaluate the acute and chronic effects of high-altitude exposure on classical sperm parameters, redox status, and membrane composition in a group of travellers. Five healthy Italian males, all lowlanders not accustomed to the altitude, were evaluated after 19 days-trekking through low, moderate, and high altitudes in the Himalayas. Sperm samples were collected before (Pre), 10 days after (Post), and 70 days after the end of the expedition (Follow-up). Sperm concentration, cholesterol and oxysterol membrane content, and redox status were measured. Hypoxic trek led to a significant reduction in sperm concentration (p < 0.001, η2p = 0.91), with a reduction from Pre to Post (71.33 ± 38.81 to 60.65 ± 34.63 × 106/mL) and a further reduction at Follow-up (to 37.13 ± 39.17 × 106/mL). The seminal volume was significantly affected by the hypoxic trek (p = 0.001, η2p = 0.75) with a significant reduction from Pre to Post (2.86 ± 0.75 to 1.68 ± 0.49 mL) and with partial recovery at Follow-up (to 2.46 ± 0.45 mL). Moreover, subjects had an increase in ROS production (+86%), and a decrease in antioxidant capacity (−37%) in the Post period with partial recovery at Follow-up. These results integrated the hormonal response on thyroid function, hypothalamus−pituitary−gonadal axis, and the prolactin/cortisol pathways previously reported. An uncontrolled ROS production, rather than a compromised antioxidant activity, was likely the cause of impaired sperm quality. The reduction in fertility status observed in this study may lie in an evolutionary Darwinian explanation, i.e., limiting reproduction due to the “adaptive disadvantage” offered by the combined stressors of high-altitude hypoxia and daily physical exercise.
Collapse
Affiliation(s)
- Vittore Verratti
- Department of Psychological, Health and Territorial Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20162 Milan, Italy
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Iva Sabovic
- Department of Medicine, University of Padova, 35122 Padova, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Via Marzolo 5, 35128 Padova, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Giorgia Scarfò
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Camillo Di Giulio
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Andrea Garolla
- Department of Medicine, University of Padova, 35122 Padova, Italy
| |
Collapse
|
31
|
Margiana R, Pakpahan C, Pangestu M. A systematic review of retinoic acid in the journey of spermatogonium to spermatozoa: From basic to clinical application. F1000Res 2022; 11:552. [PMID: 35967975 PMCID: PMC9345263 DOI: 10.12688/f1000research.110510.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Retinoic acid plays an essential role in testicular development and functions, especially spermatogenesis. We have reviewed the role of retinoic acid from basic (molecular) to clinical application. Methods: A search was conducted in the online database including PubMed, Google Scholar, and Scopus for English studies published in the last eight years about this issue. We used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines in assessing the studies we are going to investigate. Results: Studies indicated that retinoic acid plays an essential role during pluripotent stem cell migration and lineage commitment, cell differentiation, apoptosis, stem cell number regulation, and maturation arrest in spermatogenic cells. Retinoic acid can also affect related protein expression and signaling pathways at different stages of spermatogenesis. Four studies have applied retinoic acid to humans, all of them in the single-arm observational study. The results look promising but need further research with more controlled study methods, randomization, and large samples. Conclusions: This current systematic review emphasizes a novel retinoic acid mechanism that has not been well described in the literature previously on its functions during the first seven days of spermatogenesis, leading to new directions or explanations of male infertility cause and treatments as a part of reproductive health care.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Study Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Cennikon Pakpahan
- Andrology Study Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Mulyoto Pangestu
- Education Program in Reproduction and Development (EPRD), Department of Obstetrics and Gynaecology, Monash Clinical School, Monash University, Clayton, Australia
| |
Collapse
|
32
|
Ceyhan Y, Zhang M, Sandoval CG, Agoulnik AI, Agoulnik IU. Expression pattern and the roles of phosphatidylinositol phosphatases in testis. Biol Reprod 2022; 107:902-915. [PMID: 35766372 DOI: 10.1093/biolre/ioac132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/02/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Phosphoinositides (PIs) are relatively rare lipid components of the cellular membranes. Their homeostasis is tightly controlled by specific PI kinases and phosphatases. PIs play essential roles in cellular signaling, cytoskeletal organization, and secretory processes in various diseases and normal physiology. Gene targeting experiments strongly suggest that in mice with deficiency of several PI phosphatases such as Pten, Mtmrs, Inpp4b, and Inpp5b, spermatogenesis is affected, resulting in partial or complete infertility. Similarly, in men, loss of several of the PIP phosphatases is observed in infertility characterized by the lack of mature sperm. Using available gene expression databases, we compare expression of known PI phosphatases in various testicular cell types, infertility patients, and mouse age-dependent testicular gene expression, and discuss their potential roles in testis physiology and spermatogenesis.
Collapse
Affiliation(s)
- Yasemin Ceyhan
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Manqi Zhang
- Department of Medicine, Duke University, Durham, NC, USA
| | - Carlos G Sandoval
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,New York University Grossman School of Medicine, New York, NY, USA
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| | - Irina U Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
33
|
Barrachina F, de la Iglesia A, Jodar M, Soler-Ventura A, Mallofré C, Rodriguez-Carunchio L, Goudarzi A, Corral JM, Ballescà JL, Castillo J, Oliva R. Histone H4 acetylation is dysregulated in active seminiferous tubules adjacent to testicular tumours. Hum Reprod 2022; 37:1712-1726. [PMID: 35678707 DOI: 10.1093/humrep/deac130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Is histone H4 acetylation (H4ac) altered in the seminiferous tubules of patients affected by testicular tumours? SUMMARY ANSWER A considerable dysregulation of H4ac was detected in the cells of the seminiferous tubules adjacent to testicular tumours of different aetiology and prior to any treatment, while no comparable alterations were observed in patients with disrupted spermatogenesis. WHAT IS KNOWN ALREADY Altered H4ac levels have been associated with a variety of testicular pathological conditions. However, no information has been available regarding potential alterations in the spermatogenic cells adjacent to the neoplasia in testicular tumour patients. STUDY DESIGN, SIZE, DURATION A retrospective analysis using testicular sections from 33 men aged between 21 and 74 years old was performed. Three study groups were defined and subjected to double-blind evaluation: a control group with normal spermatogenesis (n = 6), patients with testicular tumours (n = 18) and patients with spermatogenic impairments (n = 8). One additional sample with normal spermatogenesis was used as a technical internal control in all evaluations. PARTICIPANTS/MATERIALS, SETTING, METHODS Immunohistochemistry against H4ac and, when needed, Placental-like alkaline phosphatase and CD117, was performed on testicular sections. The H4ac H-score, based on the percentage of detection and signal intensity, was used as the scoring method for statistical analyses. Protein expression data from the Human Protein Atlas were used to compare the expression levels of predicted secreted proteins from testicular tumours with those present in the normal tissue. MAIN RESULTS AND THE ROLE OF CHANCE We revealed, for the first time, a dramatic disruption of the spermatogenic H4ac pattern in unaffected seminiferous tubule cells from different testicular tumour patients prior to any antineoplastic treatment, as compared to controls (P < 0.05). Since no similar alterations were associated with spermatogenic impairments and the in silico analysis revealed proteins potentially secreted by the tumour to the testicular stroma, we propose a potential paracrine effect of the neoplasia as a mechanistic hypothesis for this dysregulation. LIMITATIONS, REASONS FOR CAUTION Statistical analyses were not performed on the hypospermatogenesis and Leydig cell tumour groups due to limited availability of samples. WIDER IMPLICATIONS OF THE FINDINGS To the best of our knowledge, this is the first report showing an epigenetic alteration in cells from active seminiferous tubules adjacent to tumour cells in testicular tumour patients. Our results suggest that, despite presenting spermatogenic activity, the global epigenetic dysregulation found in the testicular tumour patients could lead to molecular alterations of the male germ cells. Since testicular tumours are normally diagnosed in men at reproductive age, H4ac alterations might have an impact when these testicular tumour patients express a desire for fatherhood. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the European Union Marie Curie European Training Network actions and by grants to R.O. from the 'Ministerio de Economía y Competividad (Spain)' (fondos FEDER 'una manera de hacer Europa', PI13/00699, PI16/00346 and PI20/00936) and from EU-FP7-PEOPLE-2011-ITN289880. J.C. was supported by the Sara Borrell Postdoctoral Fellowship, Acción Estratégica en Salud, CD17/00109. J.C. is a Serra Húnter fellow (Universitat de Barcelona, Generalitat de Catalunya). F.B. has received grants from the Ministerio de Educación, Cultura y Deporte para la Formación de Profesorado Universitario (Spain) (FPU15/02306). A.d.l.I. is supported by a fellowship of the Ministerio de Economía, Industria y Competitividad (Spain) (PFIS, FI17/00224). M.J. is supported by the Government of Catalonia (Generalitat de Catalunya, pla estratègic de recerca i innovació en salut, PERIS 2016-2020, SLT002/16/00337). The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Ferran Barrachina
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain
| | - Alberto de la Iglesia
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain
| | - Meritxell Jodar
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain.,Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain
| | - Ada Soler-Ventura
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain
| | - Carme Mallofré
- Department of Pathology, Hospital Clínic, Barcelona, Spain
| | - Leonardo Rodriguez-Carunchio
- Department of Pathology, Hospital Clínic, Barcelona, Spain.,Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Barcelona, Spain
| | - Afsaneh Goudarzi
- CNRS UMR 5309, INSERM U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Juan Manuel Corral
- Department of Urology, IDIBAPS, Hospital Clínic, Barcelona, Spain.,Institute of Gynaecology, Obstetrics and Neonatology, Hospital Clínic, Barcelona, Spain
| | - Josep Lluís Ballescà
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain.,Institute of Gynaecology, Obstetrics and Neonatology, Hospital Clínic, Barcelona, Spain
| | - Judit Castillo
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Oliva
- Molecular Biology of Reproduction and Development Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain.,Biochemistry and Molecular Genetics Service, Hospital Clinic, Barcelona, Spain
| |
Collapse
|
34
|
Chen S, An G, Wang H, Wu X, Ping P, Hu L, Chen Y, Fan J, Cheng CY, Sun F. Human obstructive (postvasectomy) and nonobstructive azoospermia - Insights from scRNA-Seq and transcriptome analysis. Genes Dis 2022; 9:766-776. [PMID: 35782978 PMCID: PMC9243341 DOI: 10.1016/j.gendis.2020.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/06/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022] Open
Abstract
A substantial number of male infertility is caused by azoospermia. However, the underlying etiology and the molecular basis remain largely unknown. Through single-cell (sc)RNA sequencing, we had analyzed testis biopsy samples from two patients with obstructive azoospermia (OA) and nonobstructive azoospermia (NOA). We found only somatic cells in the NOA samples and explored the transcriptional changes in Sertoli cells in response to a loss of interactions with germ cells. Moreover, we observed a germ cell population discrepancy between an OA (postvasectomy) patient and a healthy individual. We confirmed this observation in a secondary study with two datasets at GSM3526588 and GSE124263 for detailed analysis wherein the regulatory mechanisms at the transcriptional level were identified. These findings thus provide valuable information on human spermatogenesis, and we also identified insightful information for further research on reproduction-related diseases.
Collapse
Affiliation(s)
- Shitao Chen
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, PR China
| | - Geng An
- Department of Reproductive Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, PR China
| | - Hanshu Wang
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, PR China
| | - Xiaolong Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, Jiangsu 226001, PR China
| | - Ping Ping
- Department of Urology, Shanghai Human Sperm Bank, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200120, PR China
| | - Longfei Hu
- Singleron Biotechnologies Ltd, Nanjing, Jiangsu 210000, PR China
| | - Yunmei Chen
- Singleron Biotechnologies Ltd, Nanjing, Jiangsu 210000, PR China
| | - Jue Fan
- Singleron Biotechnologies Ltd, Nanjing, Jiangsu 210000, PR China
| | - C. Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY 10065, USA
| | - Fei Sun
- International Peace Maternity and Child Health Hospital, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, PR China
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, Jiangsu 226001, PR China
| |
Collapse
|
35
|
Ruthig VA, Lamb DJ. Updates in Sertoli Cell-Mediated Signaling During Spermatogenesis and Advances in Restoring Sertoli Cell Function. Front Endocrinol (Lausanne) 2022; 13:897196. [PMID: 35600584 PMCID: PMC9114725 DOI: 10.3389/fendo.2022.897196] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Since their initial description by Enrico Sertoli in 1865, Sertoli cells have continued to enchant testis biologists. Testis size and germ cell carrying capacity are intimately tied to Sertoli cell number and function. One critical Sertoli cell function is signaling from Sertoli cells to germ cells as part of regulation of the spermatogenic cycle. Sertoli cell signals can be endocrine or paracrine in nature. Here we review recent advances in understanding the interplay of Sertoli cell endocrine and paracrine signals that regulate germ cell state. Although these findings have long-term implications for treating male infertility, recent breakthroughs in Sertoli cell transplantation have more immediate implications. We summarize the surge of advances in Sertoli cell ablation and transplantation, both of which are wedded to a growing understanding of the unique Sertoli cell niche in the transitional zone of the testis.
Collapse
Affiliation(s)
- Victor A. Ruthig
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Sexual Medicine Lab, Weill Cornell Medicine, New York, NY, United States
| | - Dolores J. Lamb
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Center for Reproductive Genomics, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Dolores J. Lamb,
| |
Collapse
|
36
|
Campolo F, Capponi C, Tarsitano MG, Tenuta M, Pozza C, Gianfrilli D, Magliocca F, Venneri MA, Vicini E, Lenzi A, Isidori AM, Barbagallo F. cAMP-specific phosphodiesterase 8A and 8B isoforms are differentially expressed in human testis and Leydig cell tumor. Front Endocrinol (Lausanne) 2022; 13:1010924. [PMID: 36277728 PMCID: PMC9585345 DOI: 10.3389/fendo.2022.1010924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
Cyclic adenosine monophosphate/Protein kinase A (cAMP/PKA) signaling pathway is the master regulator of endocrine tissue function. The level, compartmentalization and amplitude of cAMP response are finely regulated by phosphodiesterases (PDEs). PDE8 is responsible of cAMP hydrolysis and its expression has been characterized in all steroidogenic cell types in rodents including adrenal and Leydig cells in rodents however scarce data are currently available in humans. Here we demonstrate that human Leydig cells express both PDE8A and PDE8B isoforms. Interestingly, we found that the expression of PDE8B but not of PDE8A is increased in transformed Leydig cells (Leydig cell tumors-LCTs) compared to non-tumoral cells. Immunofluorescence analyses further reveals that PDE8A is also highly expressed in specific spermatogenic stages. While the protein is not detected in spermatogonia it accumulates nearby the forming acrosome, in the trans-Golgi apparatus of spermatocytes and spermatids and it follows the fate of this organelle in the later stages translocating to the caudal part of the cell. Taken together our findings suggest that 1) a specific pool(s) of cAMP is/are regulated by PDE8A during spermiogenesis pointing out a possible new role of this PDE8 isoform in key events governing the differentiation and maturation of human sperm and 2) PDE8B can be involved in Leydig cell transformation.
Collapse
Affiliation(s)
- Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Chiara Capponi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Maria Grazia Tarsitano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Marta Tenuta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Magliocca
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Mary A. Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena Vicini
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea M. Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Faculty of Medicine and Surgery, Kore University of Enna, Enna, Italy
- *Correspondence: Federica Barbagallo,
| |
Collapse
|
37
|
Huang ZH, Huang C, Ji XR, Zhou WJ, Luo XF, Liu Q, Tang YL, Gong F, Zhu WB. MKK7-mediated phosphorylation of JNKs regulates the proliferation and apoptosis of human spermatogonial stem cells. World J Stem Cells 2021; 13:1797-1812. [PMID: 34909124 PMCID: PMC8641020 DOI: 10.4252/wjsc.v13.i11.1797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/28/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human spermatogonial stem cells (SSCs) are the basis of spermatogenesis. However, little is known about the developmental regulatory mechanisms of SSC due to sample origin and species differences.
AIM To investigates the mechanisms involved in the proliferation of human SSC.
METHODS The expression of mitogen-activated protein kinase kinase 7 (MKK7) in human testis was identified using immunohistochemistry and western blotting (WB). MKK7 was knocked down using small interfering RNA, and cell proliferation and apoptosis were detected by WB, EdU, cell counting kit-8 and fluorescence-activated cell sorting. After bioinformatic analysis, the interaction of MKK7 with c-Jun N-terminal kinases ( JNKs ) was verified by protein co-immunoprecipitation and WB. The phosphorylation of JNKs was inhibited by SP600125, and the phenotypic changes were detected by WB, cell counting kit-8 and fluorescence-activated cell sorting.
RESULTS MKK7 is mainly expressed in human SSCs, and MKK7 knockdown inhibits SSC proliferation and promotes their apoptosis. MKK7 mediated the phosphorylation of JNKs, and after inhibiting the phosphorylation of JNKs, the phenotypic changes of the cells were similar to those after MKK7 downregulation. The expression of MKK7 was significantly downregulated in patients with abnormal spermatogenesis, suggesting that abnormal MKK7 may be associated with spermatogenesis impairment.
CONCLUSION MKK7 regulates the proliferation and apoptosis of human SSC by mediating the phosphorylation of JNKs.
Collapse
Affiliation(s)
- Zeng-Hui Huang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
- Department of Reproductive Center, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Chuan Huang
- Department of Sperm Bank, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Xi-Ren Ji
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Jun Zhou
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Xue-Feng Luo
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Liu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Yu-Lin Tang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Bing Zhu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
38
|
de Nie I, Mulder CL, Meißner A, Schut Y, Holleman EM, van der Sluis WB, Hannema SE, den Heijer M, Huirne J, van Pelt AMM, van Mello NM. Histological study on the influence of puberty suppression and hormonal treatment on developing germ cells in transgender women. Hum Reprod 2021; 37:297-308. [PMID: 34791270 PMCID: PMC8804334 DOI: 10.1093/humrep/deab240] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022] Open
Abstract
STUDY QUESTION Can transgender women cryopreserve germ cells obtained from their orchiectomy specimen for fertility preservation, after having used puberty suppression and/or hormonal treatment? SUMMARY ANSWER In the vast majority of transgender women, there were still immature germ cells present in the orchiectomy specimen, and in 4.7% of transgender women—who all initiated medical treatment in Tanner stage 4 or higher—mature spermatozoa were found, which would enable cryopreservation of spermatozoa or testicular tissue after having used puberty suppression and/or hormonal treatment. WHAT IS KNOWN ALREADY Gender affirming treatment (i.e. puberty suppression, hormonal treatment, and subsequent orchiectomy) impairs reproductive function in transgender women. Although semen cryopreservation is generally offered during the transition process, this option is not feasible for all transgender women (e.g. due to incomplete spermatogenesis when initiating treatment in early puberty, in case of inability to masturbate, or when temporary cessation of hormonal treatment is too disruptive). Harvesting mature spermatozoa, or testicular tissue harboring immature germ cells, from orchiectomy specimens obtained during genital gender-affirming surgery (gGAS) might give this group a chance of having biological children later in life. Previous studies on spermatogenesis in orchiectomy specimens showed conflicting results, ranging from complete absence of germ cells to full spermatogenesis, and did not involve transgender women who initiated medical treatment in early- or late puberty. STUDY DESIGN, SIZE, DURATION Histological and immunohistochemical analyses were performed on orchiectomy specimens from 214 transgender women who underwent gGAS between 2006 and 2018. Six subgroups were identified, depending on pubertal stage at initiation of medical treatment (Tanner stage 2-3, Tanner stage 4-5, adult), and whether hormonal treatment was continued or temporarily stopped prior to gGAS in each of these groups. PARTICIPANTS/MATERIALS, SETTING, METHODS All transgender women used a combination of estrogens and testosterone suppressing therapy. Orchiectomy specimen sections were stained with Mayer’s hematoxylin and eosin and histologically analyzed to assess the Johnsen score and the ratio of most advanced germ cell types in at least 50 seminiferous tubular cross-sections. Subsequently, immunohistochemistry was used to validate these findings using spermatogonia, spermatocytes or spermatids markers (MAGE-A3/A4, γH2AX, Acrosin, respectively). Possibilities for fertility preservation were defined as: preservation of spermatozoa, preservation of spermatogonial stem cells or no possibilities (in case no germ cells were found). Outcomes were compared between subgroups and logistic regression analyses were used to assess the association between the duration of hormonal treatment and the possibilities for fertility preservation. MAIN RESULTS AND THE ROLE OF CHANCE Mature spermatozoa were encountered in 4.7% of orchiectomy specimens, all from transgender women who had initiated medical treatment in Tanner stage 4 or higher. In 88.3% of the study sample orchiectomy specimens only contained immature germ cells (round spermatids, spermatocytes or spermatogonia, as most advanced germ cell type). In 7.0%, a complete absence of germ cells was observed, all these samples were from transgender women who had initiated medical treatment in adulthood. Cessation of hormonal treatment prior to gGAS did not affect the presence of germ cells or their maturation stage, nor was there an effect of the duration of hormonal treatment prior to gGAS. LIMITATIONS, REASONS FOR CAUTION Since data on serum hormone levels on the day of gGAS were not available, we were unable to verify if the transgender women who were asked to temporarily stop hormonal treatment 4 weeks prior to surgery actually did so, and if people with full spermatogenesis were compliant to treatment. WIDER IMPLICATIONS OF THE FINDINGS There may still be options for fertility preservation in orchiectomy specimens obtained during gGAS since a small percentage of transgender women had full spermatogenesis, which could enable cryopreservation of mature spermatozoa via a testicular sperm extraction procedure. Furthermore, the vast majority still had immature germ cells, which could enable cryopreservation of testicular tissue harboring spermatogonial stem cells. If maturation techniques like in vitro spermatogenesis become available in the future, harvesting germ cells from orchiectomy specimens might be a promising option for those who are otherwise unable to have biological children. STUDY FUNDING/COMPETING INTEREST None. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- I de Nie
- Department of Endocrinology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Obstetrics and Gynecology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C L Mulder
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - A Meißner
- Department of Endocrinology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Obstetrics and Gynecology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Y Schut
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - E M Holleman
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - W B van der Sluis
- Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S E Hannema
- Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M den Heijer
- Department of Endocrinology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Huirne
- Department of Obstetrics and Gynecology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - A M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - N M van Mello
- Center of Expertise on Gender Dysphoria, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Obstetrics and Gynecology, Amsterdam UMC, Amsterdam Reproduction & Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Subrini J, Turner J. Y chromosome functions in mammalian spermatogenesis. eLife 2021; 10:67345. [PMID: 34606444 PMCID: PMC8489898 DOI: 10.7554/elife.67345] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
The mammalian Y chromosome is critical for male sex determination and spermatogenesis. However, linking each Y gene to specific aspects of male reproduction has been challenging. As the Y chromosome is notoriously hard to sequence and target, functional studies have mostly relied on transgene-rescue approaches using mouse models with large multi-gene deletions. These experimental limitations have oriented the field toward the search for a minimum set of Y genes necessary for male reproduction. Here, considering Y-chromosome evolutionary history and decades of discoveries, we review the current state of research on its function in spermatogenesis and reassess the view that many Y genes are disposable for male reproduction.
Collapse
Affiliation(s)
- Jeremie Subrini
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - James Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
40
|
Zhou D, Fan J, Liu Z, Tang R, Wang X, Bo H, Zhu F, Zhao X, Huang Z, Xing L, Tao K, Zhang H, Nie H, Zhang H, Zhu W, He Z, Fan L. TCF3 Regulates the Proliferation and Apoptosis of Human Spermatogonial Stem Cells by Targeting PODXL. Front Cell Dev Biol 2021; 9:695545. [PMID: 34422820 PMCID: PMC8377737 DOI: 10.3389/fcell.2021.695545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are the initial cells for the spermatogenesis. Although much progress has been made on uncovering a number of modulators for the SSC fate decisions in rodents, the genes mediating human SSCs remain largely unclear. Here we report, for the first time, that TCF3, a member of the basic helix-loop-helix family of transcriptional modulator proteins, can stimulate proliferation and suppress the apoptosis of human SSCs through targeting podocalyxin-like protein (PODXL). TCF3 was expressed primarily in GFRA1-positive spermatogonia, and EGF (epidermal growth factor) elevated TCF3 expression level. Notably, TCF3 enhanced the growth and DNA synthesis of human SSCs, whereas it repressed the apoptosis of human SSCs. RNA sequencing and chromatin immunoprecipitation (ChIP) assays revealed that TCF3 protein regulated the transcription of several genes, including WNT2B, TGFB3, CCN4, MEGF6, and PODXL, while PODXL silencing compromised the stem cell activity of SSCs. Moreover, the level of TCF3 protein was remarkably lower in patients with spermatogenesis failure when compared to individuals with obstructive azoospermia with normal spermatogenesis. Collectively, these results implicate that TCF3 modulates human SSC proliferation and apoptosis through PODXL. This study is of great significance since it would provide a novel molecular mechanism underlying the fate determinations of human SSCs and it could offer new targets for gene therapy of male infertility.
Collapse
Affiliation(s)
- Dai Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.,College of Life Sciences, Hunan Normal University, Changsha, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China.,NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
| | - Jingyu Fan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States
| | - Zhizhong Liu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Department of Urology, Hunan Cancer Hospital, Changsha, China
| | - Ruiling Tang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Xingming Wang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Hao Bo
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Fang Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Xueheng Zhao
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Zenghui Huang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Liu Xing
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Ke Tao
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.,The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Han Zhang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Hongchuan Nie
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Huan Zhang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Wenbing Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Liqing Fan
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China.,NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
| |
Collapse
|
41
|
Wu X, Gao S, Wang L, Bu T, Wu S, Zhou L, Shi J, Wu D, Sun F, Cheng CY. Role of laminin and collagen chains in human spermatogenesis - Insights from studies in rodents and scRNA-Seq transcriptome profiling. Semin Cell Dev Biol 2021; 121:125-132. [PMID: 34325997 DOI: 10.1016/j.semcdb.2021.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022]
Abstract
Studies have demonstrated that biologically active fragments are generated from the basement membrane and the Sertoli cell-spermatid adhesion site known as apical ectoplasmic specialization (apical ES, a testis-specific actin-based anchoring junction) in the rat testis. These bioactive fragments or peptides are produced locally across the seminiferous epithelium through proteolytic cleavage of constituent proteins at the basement membrane and the apical ES. Studies have shown that they are being used to modulate and coordinate cellular functions across the seminiferous epithelium during different stages of the epithelial cycle of spermatogenesis. In this review, we briefly summarize recent findings based on studies using rat testes as a study model regarding the role of these bioactive peptides that serve as a local regulatory network to support spermatogenesis. We also used scRNA-Seq transcriptome datasets in the public domain for OA (obstructive azoospermia) and NAO (non-obstructive azoospermia) human testes versus testes from normal men for analysis in this review. It was shown that there are differential expression of different collagen chains and laminin chains in these testes, suggesting the possibility of a similar local regulatory network in the human testis to support spermatogenesis, and the possible disruption of such network in men is associated with OA and/or NOA.
Collapse
Affiliation(s)
- Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Sheng Gao
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Tiao Bu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Siwen Wu
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Liwei Zhou
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Jie Shi
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Di Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China.
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA.
| |
Collapse
|
42
|
Azhar M, Altaf S, Uddin I, Cheng J, Wu L, Tong X, Qin W, Bao J. Towards Post-Meiotic Sperm Production: Genetic Insight into Human Infertility from Mouse Models. Int J Biol Sci 2021; 17:2487-2503. [PMID: 34326689 PMCID: PMC8315030 DOI: 10.7150/ijbs.60384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Declined quality and quantity of sperm is currently the major cause of patients suffering from infertility. Male germ cell development is spatiotemporally regulated throughout the whole developmental process. While it has been known that exogenous factors, such as environmental exposure, diet and lifestyle, et al, play causative roles in male infertility, recent progress has revealed abundant genetic mutations tightly associated with defective male germline development. In mammals, male germ cells undergo dramatic morphological change (i.e., nuclear condensation) and chromatin remodeling during post-meiotic haploid germline development, a process termed spermiogenesis; However, the molecular machinery players and functional mechanisms have yet to be identified. To date, accumulated evidence suggests that disruption in any step of haploid germline development is likely manifested as fertility issues with low sperm count, poor sperm motility, aberrant sperm morphology or combined. With the continually declined cost of next-generation sequencing and recent progress of CRISPR/Cas9 technology, growing studies have revealed a vast number of disease-causing genetic variants associated with spermiogenic defects in both mice and humans, along with mechanistic insights partially attained and validated through genetically engineered mouse models (GEMMs). In this review, we mainly summarize genes that are functional at post-meiotic stage. Identification and characterization of deleterious genetic variants should aid in our understanding of germline development, and thereby further improve the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Saba Altaf
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Islam Uddin
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Jinbao Cheng
- The 901th hospital of Joint logistics support Force of PLA, Anhui, China
| | - Limin Wu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Xianhong Tong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| |
Collapse
|
43
|
Vereecke G, Defreyne J, Van Saen D, Collet S, Van Dorpe J, T'Sjoen G, Goossens E. Characterisation of testicular function and spermatogenesis in transgender women. Hum Reprod 2021; 36:5-15. [PMID: 33257947 DOI: 10.1093/humrep/deaa254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/08/2020] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Does gender-affirming treatment prevent full spermatogenesis in transgender women (TW)? SUMMARY ANSWER Adequate hormonal therapy (HT) leads to complete suppression of spermatogenesis in most TW, if serum testosterone levels within female reference ranges are obtained. WHAT IS KNOWN ALREADY Gender-affirming treatment in transgender individuals may involve gender-affirming HT. The effects on spermatogenesis in TW remain unclear. In order to add information from a referral centre for transgender care, we wish to compare results of earlier studies with our population of TW who received a standard hormone treatment. STUDY DESIGN, SIZE, DURATION This was a prospective cohort study part of the European Network for the Investigation of Gender Incongruence (ENIGI), conducted between 15 February 2010 and 30 September 2015. There were 162 TW were included in the ENIGI study at the Ghent University Hospital in Belgium. Participants are included in ENIGI when they first start HT, and follow-up visits occur over the next 3 years. PARTICIPANTS/MATERIALS, SETTING METHODS The study included 97 TW who initiated HT with cyproterone acetate (CPA) plus oestrogens and proceeded with gonadectomy at the Ghent University Hospital. Testicular tissue retrieved during gonadectomy was processed and stained for four different germ cell markers by the Biology of the Testis lab at the Vrije Universiteit Brussel. Subsequent immunohistochemical staining was performed for melanoma-associated antigen A4 (MAGE-A4, marker for spermatogonia and early spermatocytes), boule homologue, RNA-binding protein (BOLL, marker for secondary spermatocytes and round spermatids), cAMP-responsive element modulator (CREM, marker for round spermatids) and acrosin (marker for acrosome visualization). Serum levels of sex steroids were measured prior to surgery. MAIN RESULTS AND THE ROLE OF CHANCE Suppressed testosterone levels (<50 ng/dl) were found in 92% of the participants prior to surgery. The mean time between initiation of HT and surgery was 685 days. In 88% (85/97) of the sections, MAGE-A4 staining was positive. Further staining could not reveal complete spermatogenesis in any participant. LIMITATIONS, REASONS FOR CAUTION Testicular function of the participants prior to initiation of HT was not assessed, although all participants presented with cisgender male serum testosterone values before initiation of HT. The current study only reports on people using CPA at a fixed dose and may therefore not be applicable to all TW. WIDER IMPLICATIONS OF THE FINDINGS HT leads to complete suppression of spermatogenesis in most TW, if serum testosterone levels within female reference ranges are obtained. Serum testosterone levels are associated with the sperm maturation rate. It is important to discuss sperm preservation before the start of hormone therapy. If serum testosterone levels remain higher, spermatogenesis may still occur. STUDY FUNDING/COMPETING INTEREST(S) D.V.S. is a post-doctoral fellow of the Fonds Wetenschappelijk Onderzoek (FWO; 12M2819N). Processing of the testis specimens was funded by the Biology of The Testes (BITE) research group (Department of Reproduction, Genetics and Regenerative medicine at Vrije Universiteit Brussel (VUB)). There are no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Gertjan Vereecke
- Department of Endocrinology, AZ Groeninge, 8500 Kortrijk, Belgium
| | - Justine Defreyne
- Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Dorien Van Saen
- Department of Genetics and Regenerative Medicine, Biology of the Testis, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Sarah Collet
- Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Guy T'Sjoen
- Department of Endocrinology and Center for Sexology and Gender, Ghent University Hospital, 9000 Ghent, Belgium
| | - Ellen Goossens
- Department of Genetics and Regenerative Medicine, Biology of the Testis, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
44
|
Li H, Liu S, Wu S, Li L, Ge R, Cheng CY. Bioactive fragments of laminin and collagen chains: lesson from the testis. Reproduction 2021; 159:R111-R123. [PMID: 31581125 DOI: 10.1530/rep-19-0288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022]
Abstract
Recent studies have shown that the testis is producing several biologically active peptides, namely the F5- and the NC1-peptides from laminin-γ3 and collagen α3 (IV) chain, respectively, that promotes blood-testis barrier (BTB) remodeling and also elongated spermatid release at spermiation. Also the LG3/4/5 peptide from laminin-α2 chain promotes BTB integrity which is likely being used for the assembly of a 'new' BTB behind preleptotene spermatocytes under transport at the immunological barrier. These findings thus provide a new opportunity for investigators to better understand the biology of spermatogenesis. Herein, we briefly summarize the recent findings and provide a critical update. We also present a hypothetical model which could serve as the framework for studies in the years to come.
Collapse
Affiliation(s)
- Huitao Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Shiwen Liu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Linxi Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Renshan Ge
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| |
Collapse
|
45
|
Mularoni V, Esposito V, Di Persio S, Vicini E, Spadetta G, Berloco P, Fanelli F, Mezzullo M, Pagotto U, Pelusi C, Nielsen JE, Rajpert-De Meyts E, Jorgensen N, Jorgensen A, Boitani C. Age-related changes in human Leydig cell status. Hum Reprod 2021; 35:2663-2676. [PMID: 33094328 DOI: 10.1093/humrep/deaa271] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
STUDY QUESTION What are the consequences of ageing on human Leydig cell number and hormonal function? SUMMARY ANSWER Leydig cell number significantly decreases in parallel with INSL3 expression and Sertoli cell number in aged men, yet the in vitro Leydig cell androgenic potential does not appear to be compromised by advancing age. WHAT IS KNOWN ALREADY There is extensive evidence that ageing is accompanied by decline in serum testosterone levels, a general involution of testis morphology and reduced spermatogenic function. A few studies have previously addressed single features of the human aged testis phenotype one at a time, but mostly in tissue from patients with prostate cancer. STUDY DESIGN, SIZE, DURATION This comprehensive study examined testis morphology, Leydig cell and Sertoli cell number, steroidogenic enzyme expression, INSL3 expression and androgen secretion by testicular fragments in vitro. The majority of these endpoints were concomitantly evaluated in the same individuals that all displayed complete spermatogenesis. PARTICIPANTS/MATERIALS, SETTING, METHODS Testis biopsies were obtained from 15 heart beating organ donors (age range: 19-85 years) and 24 patients (age range: 19-45 years) with complete spermatogenesis. Leydig cells and Sertoli cells were counted following identification by immunohistochemical staining of specific cell markers. Gene expression analysis of INSL3 and steroidogenic enzymes was carried out by qRT-PCR. Secretion of 17-OH-progesterone, dehydroepiandrosterone, androstenedione and testosterone by in vitro cultured testis fragments was measured by LC-MS/MS. All endpoints were analysed in relation to age. MAIN RESULTS AND THE ROLE OF CHANCE Increasing age was negatively associated with Leydig cell number (R = -0.49; P < 0.01) and concomitantly with the Sertoli cell population size (R= -0.55; P < 0.001). A positive correlation (R = 0.57; P < 0.001) between Sertoli cell and Leydig cell numbers was detected at all ages, indicating that somatic cell attrition is a relevant cellular manifestation of human testis status during ageing. INSL3 mRNA expression (R= -0.52; P < 0.05) changed in parallel with Leydig cell number and age. Importantly, steroidogenic capacity of Leydig cells in cultured testis tissue fragments from young and old donors did not differ. Consistently, age did not influence the mRNA expression of steroidogenic enzymes. The described changes in Leydig cell phenotype with ageing are strengthened by the fact that the different age-related effects were mostly evaluated in tissue from the same men. LIMITATIONS, REASONS FOR CAUTION In vitro androgen production analysis could not be correlated with in vivo hormone values of the organ donors. In addition, the number of samples was relatively small and there was scarce information about the concomitant presence of potential confounding variables. WIDER IMPLICATIONS OF THE FINDINGS This study provides a novel insight into the effects of ageing on human Leydig cell status. The correlation between Leydig cell number and Sertoli cell number at any age implies a connection between these two cell types, which may be of particular relevance in understanding male reproductive disorders in the elderly. However aged Leydig cells do not lose their in vitro ability to produce androgens. Our data have implications in the understanding of the physiological role and regulation of intratesticular sex steroid levels during the complex process of ageing in humans. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from Prin 2010 and 2017. The authors have no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Valentina Mularoni
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopedic Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| | - Valentina Esposito
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopedic Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| | - Sara Di Persio
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopedic Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| | - Elena Vicini
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopedic Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| | - Gustavo Spadetta
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiological and Geriatric Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| | - Pasquale Berloco
- Department of General and Specialistic Surgery "Paride Stefanini", University of Rome "La Sapienza", 00161 Rome, Italy
| | - Flaminia Fanelli
- Endocrinology and Diabetes Prevention and Care-Unit, Department of Medical and Surgical Sciences, Centre for Applied Biomedical Research, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Marco Mezzullo
- Endocrinology and Diabetes Prevention and Care-Unit, Department of Medical and Surgical Sciences, Centre for Applied Biomedical Research, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Uberto Pagotto
- Endocrinology and Diabetes Prevention and Care-Unit, Department of Medical and Surgical Sciences, Centre for Applied Biomedical Research, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - Carla Pelusi
- Endocrinology and Diabetes Prevention and Care-Unit, Department of Medical and Surgical Sciences, Centre for Applied Biomedical Research, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | - John E Nielsen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet 2100, Denmark, Copenhagen
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet 2100, Denmark, Copenhagen
| | - Niels Jorgensen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet 2100, Denmark, Copenhagen
| | - Anne Jorgensen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet 2100, Denmark, Copenhagen
| | - Carla Boitani
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopedic Sciences, University of Rome "La Sapienza", 00161 Rome, Italy
| |
Collapse
|
46
|
Kawamura M, Sugihara K, Takigawa-Imamura H, Ogawa T, Miura T. Mathematical Modeling of Dynamic Cellular Association Patterns in Seminiferous Tubules. Bull Math Biol 2021; 83:33. [PMID: 33594605 DOI: 10.1007/s11538-021-00863-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/21/2021] [Indexed: 11/29/2022]
Abstract
In vertebrates, sperm is generated in testicular tube-like structures called seminiferous tubules. The differentiation stages of spermatogenesis exhibit a dynamic spatiotemporal wavetrain pattern. There are two types of pattern-the vertical type, which is observed in mice, and the helical type, which is observed in humans. The mechanisms of this pattern difference remain little understood. In the present study, we used a three-species reaction-diffusion model to reproduce the wavetrain pattern observed in vivo. We hypothesized that the wavelength of the pattern in mice was larger than that in humans and undertook numerical simulations. We found complex patterns of helical and vertical pattern frequency, which can be understood by pattern selection using boundary conditions. From these theoretical results, we predicted that a small number of vertical patterns should be present in human seminiferous tubules. We then found vertical patterns in histological sections of human tubules, consistent with the theoretical prediction. Finally, we showed that the previously reported irregularity of the human pattern could be reproduced using two factors: a wider unstable wavenumber range and the irregular geometry of human compared with mouse seminiferous tubules. These results show that mathematical modeling is useful for understanding the pattern dynamics of seminiferous tubules in vivo.
Collapse
Affiliation(s)
- Mari Kawamura
- Academic Society of Mathematical Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | - Kei Sugihara
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hisako Takigawa-Imamura
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiyuki Ogawa
- Meiji Institute for Advanced Study of Mathematical Sciences, Meiji University, Tokyo, Japan
| | - Takashi Miura
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
47
|
Frias S, Van Hummelen P, Meistrich ML, Wyrobek AJ. Meiotic susceptibility for induction of sperm with chromosomal aberrations in patients receiving combination chemotherapy for Hodgkin lymphoma. PLoS One 2020; 15:e0242218. [PMID: 33370316 PMCID: PMC7769287 DOI: 10.1371/journal.pone.0242218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/28/2020] [Indexed: 11/19/2022] Open
Abstract
Improvements in survival rates with gonad-sparing protocols for childhood and adolescence cancer have increased the optimism of survivors to become parents after treatment. Findings in rodents indicate that chromosomal aberrations can be induced in male germ cells by genotoxic exposures and transmitted to offspring and future generations with effects on development, fertility and health. Thus, there is a need for effective technologies to identify human sperm carrying chromosomal aberrations to assess the germ-line risks, especially for cancer survivors who have received genotoxic therapies. The time-dependent changes in the burden of sperm carrying structural chromosomal aberrations were assessed for the first time in a cancer setting, using the AM8 sperm FISH protocol which simultaneously detects abnormalities in chromosomal structure and number in sperm. Nine Hodgkin lymphoma (HL) patients provided 20 semen samples before, during, and after NOVP therapy (Novantrone, Oncovin, Velban and Prednisone) and radiation therapy that produced scattered gonadal doses from <0.05 to 0.6 Gy. Late meiosis was found to be the most sensitive to NOVP treatment for the production of sperm with chromosomal abnormalities, both in structure and number. Earlier stages of spermatogenesis were less sensitive and there was no evidence that therapy-exposed stem cells resulted in increased frequencies of sperm with abnormalities in chromosomal structure or number. This indicates that NOVP therapy may increase the risks for paternal transmission of chromosomal structural aberrations for sperm produced 32 to 45 days after a treatment with these drugs and implies that there are no excess risks for pregnancies conceived more than 6 months after this therapy. This clinical evaluation of the AM8 sperm FISH protocol indicates that it is a promising tool for assessing an individual's burden of sperm carrying chromosomal structural aberrations as well as aneuploidies after cancer therapy, with broad applications in other clinical and environmental situations that may pose aneugenic or clastogenic risks to human spermatogenesis.
Collapse
Affiliation(s)
- Sara Frias
- Health Effects Genetics Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- Laboratorio de Citogenética, Instituto Nacional de Pediatría /Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paul Van Hummelen
- Health Effects Genetics Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- Department of Medicine, Stanford University, Palo Alto, California, United States of America
| | - Marvin L. Meistrich
- Department of Experimental Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Andrew J. Wyrobek
- Health Effects Genetics Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Fertility Preservation in Childhood Cancer: Endocrine Activity in Prepubertal Human Testis Xenografts Exposed to a Pubertal Hormone Environment. Cancers (Basel) 2020; 12:cancers12102830. [PMID: 33008013 PMCID: PMC7600569 DOI: 10.3390/cancers12102830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Substantial strides have been made in treating childhood cancers; however, as a result of chemotherapy and radiotherapy, young males experience long-term side effects, including impaired fertility. Whilst prepubertal testicular tissue can be cryopreserved prior to gonadotoxic treatments, it remains to be determined how to generate mature gametes from the immature human testis tissue. Development of immature germ cells into sperm is a complex process, which is supported by mature Sertoli cells and testosterone produced from Leydig cells. We used an established testicular xenotransplantation model to investigate the effect of puberty hormones, known as gonadotrophins, on functional maturation of the spermatogonial stem cell (SSC) niche. Limited testosterone production and partial maturation of Sertoli cells occurred in prepubertal testis grafts, suggesting that longer periods of grafting and/or identification of additional factors are required to develop testicular transplantation as a model for fertility preservation in male survivors of childhood cancer. Abstract Survivors of childhood cancer are at risk for long-term treatment-induced health sequelae, including gonadotoxicity and iatrogenic infertility. At present, for prepubertal boys there are no viable clinical options to preserve future reproductive potential. We investigated the effect of a pubertal induction regimen with gonadotrophins on prepubertal human testis xenograft development. Human testis tissue was obtained from patients with cancer and non-malignant haematological disorders (n = 6; aged 1–14 years) who underwent testis tissue cryopreservation for fertility preservation. Fresh and frozen-thawed testis fragments were transplanted subcutaneously or intratesticularly into immunocompromised mice. Graft-bearing mice received injections of vehicle or exogenous gonadotrophins, human chorionic gonadotrophin (hCG, 20 IU), and follicle-stimulating hormone (FSH, 12.5 IU) three times a week for 12 weeks. The gross morphology of vehicle and gonadotrophin-exposed grafts was similar for both transplantation sites. Exposure of prepubertal human testis tissue xenografts to exogenous gonadotrophins resulted in limited endocrine function of grafts, as demonstrated by the occasional expression of the steroidogenic cholesterol side-chain cleavage enzyme (CYP11A1). Plasma testosterone concentrations (0.13 vs. 0.25 ng/mL; p = 0.594) and seminal vesicle weights (10.02 vs. 13.93 mg; p = 0.431) in gonadotrophin-exposed recipient mice were comparable to vehicle-exposed controls. Regardless of the transplantation site and treatment, initiation and maintenance of androgen receptor (AR) expression were observed in Sertoli cells, indicating commitment towards a more differentiated status. However, neither exogenous gonadotrophins (in castrated host mice) nor endogenous testosterone (in intact host mice) were sufficient to repress the expression of markers associated with immature Sertoli cells, such as anti-Müllerian hormone (AMH) and Ki67, or to induce the redistribution of junctional proteins (connexin 43, CX43; claudin 11, CLDN11) to areas adjacent to the basement membrane. Spermatogonia did not progress developmentally but remained the most advanced germ cell type in testis xenografts. Overall, these findings demonstrate that exogenous gonadotrophins promote partial activation and maturation of the somatic environment in prepubertal testis xenografts. However, alternative hormone regimens or additional factors for pubertal induction are required to complete the functional maturation of the spermatogonial stem cell (SSC) niche.
Collapse
|
49
|
Effect of vitamin E administered to men in infertile couples on sperm and assisted reproduction outcomes: a double-blind randomized study. F S Rep 2020; 1:219-226. [PMID: 34223248 PMCID: PMC8244364 DOI: 10.1016/j.xfre.2020.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/13/2020] [Indexed: 11/24/2022] Open
Abstract
Objective To evaluate the influence on sperm parameters and in vitro fertilization (IVF) outcomes of the administration of 400 mg/day of vitamin E for 3 months to men from infertile couples who are undergoing IVF. Design Double-blind, placebo-controlled, randomized study. Setting Human reproduction unit of a university hospital. Patients A total of 101 couples, 50 in the vitamin E group and 51 in the placebo group, undergoing IVF, among whom 64.4% of cases had an abnormal spermiogram according to World Health Organization (WHO) criteria. Interventions Vitamin E (α-tocopherol), 400 mg daily by mouth for 3 months, with sperm analysis performed immediately before starting the treatment and 3 months later on the day of IVF. Main Outcome Measures WHO sperm parameters and IVF outcomes. Results Although there was a statistically significant increase in progressive motility in the vitamin E group compared with before-treatment values, a similar increase occurred in the placebo group. Normal morphology was even better in the placebo group. Regarding IVF outcomes, better fertilization rates were observed in the placebo group, but the live-birth rate per transfer was statistically significantly higher in the vitamin E group: 17 (41.46%) of 41 versus 9 (20.46%) of 44 in the placebo group. Although the clinical pregnancy rates (both per transfer and per cycle started) and the implantation rate were somewhat higher in the vitamin E group (43.9% and 25%; 36.0% and 22.0%; and 24.7% and 14.1%, respectively), the increase was not statistically significant. Conclusions The effect of vitamin E on classic sperm parameters was not an improvement over placebo. Nonetheless, vitamin E administration was associated with a statistically significantly higher live-birth rate, and there was a trend toward better results in other IVF parameters. Clinical Trial Registration Number Eudra CT 2007-000960-25.
Collapse
|
50
|
Torres-Flores U, Hernández-Hernández A. The Interplay Between Replacement and Retention of Histones in the Sperm Genome. Front Genet 2020; 11:780. [PMID: 32765595 PMCID: PMC7378789 DOI: 10.3389/fgene.2020.00780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
The genome of eukaryotes is highly organized within the cell nucleus, this organization per se elicits gene regulation and favors other mechanisms like cell memory throughout histones and their post-translational modifications. In highly specialized cells, like sperm, the genome is mostly organized by protamines, yet a significant portion of it remains organized by histones. This protamine-histone-DNA organization, known as sperm epigenome, is established during spermiogenesis. Specific histones and their post-translational modifications are retained at specific genomic sites and during embryo development these sites recapitulate their histone profile that harbored in the sperm nucleus. It is known that histones are the conduit of epigenetic memory from cell to cell, hence histones in the sperm epigenome may have a role in transmitting epigenetic memory from the sperm to the embryo. However, the exact function and mechanism of histone retention remains elusive. During spermatogenesis, most of the histones that organize the genome are replaced by protamines and their retention at specific regions may be deeply intertwined with the eviction and replacement mechanism. In this review we will cover some relevant aspects of histone replacement that in turn may help us to contextualize histone retention. In the end, we focus on the architectonical protein CTCF that is, so far, the only factor that has been directly linked to the histone retention process.
Collapse
Affiliation(s)
- Ulises Torres-Flores
- Biología de Células Individuales (BIOCELIN), Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Abrahan Hernández-Hernández
- Biología de Células Individuales (BIOCELIN), Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|