1
|
Podgrajsek R, Hodzic A, Stimpfel M, Kunej T, Peterlin B. Insight into the complexity of male infertility: a multi-omics review. Syst Biol Reprod Med 2024; 70:73-90. [PMID: 38517373 DOI: 10.1080/19396368.2024.2317804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/06/2024] [Indexed: 03/23/2024]
Abstract
Male infertility is a reproductive disorder, accounting for 40-50% of infertility. Currently, in about 70% of infertile men, the cause remains unknown. With the introduction of novel omics and advancement in high-throughput technology, potential biomarkers are emerging. The main purpose of our work was to overview different aspects of omics approaches in association with idiopathic male infertility and highlight potential genes, transcripts, non-coding RNA, proteins, and metabolites worth further exploring. Using the Gene Ontology (GO) analysis, we aimed to compare enriched GO terms from each omics approach and determine their overlapping. A PubMed database screening for the literature published between February 2014 and June 2022 was performed using the keywords: male infertility in association with different omics approaches: genomics, epigenomics, transcriptomics, ncRNAomics, proteomics, and metabolomics. A GO enrichment analysis was performed using the Enrichr tool. We retrieved 281 global studies: 171 genomics (DNA level), 21 epigenomics (19 of methylation and two histone residue modifications), 15 transcriptomics, 31 non-coding RNA, 29 proteomics, two protein posttranslational modification, and 19 metabolomics studies. Gene ontology comparison showed that different omics approaches lead to the identification of different molecular factors and that the corresponding GO terms, obtained from different omics approaches, do not overlap to a larger extent. With the integration of novel omics levels into the research of idiopathic causes of male infertility, using multi-omic systems biology approaches, we will be closer to finding the potential biomarkers and consequently becoming aware of the entire spectrum of male infertility, their cause, prognosis, and potential treatment.
Collapse
Affiliation(s)
- Rebeka Podgrajsek
- Department of Human Reproduction, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Alenka Hodzic
- Clinical Institute of Genomic Medicine, University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Novo mesto, Novo Mesto, Slovenia
| | - Martin Stimpfel
- Department of Human Reproduction, University Medical Center Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Kunej
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia
| | - Borut Peterlin
- Clinical Institute of Genomic Medicine, University Medical Center Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
2
|
Wang X, Shen G, Yang Y, Jiang C, Ruan T, Yang X, Zhuo L, Zhang Y, Ou Y, Zhao X, Long S, Tang X, Lin T, Shen Y. DNAH3 deficiency causes flagellar inner dynein arm loss and male infertility in humans and mice. eLife 2024; 13:RP96755. [PMID: 39503742 PMCID: PMC11540302 DOI: 10.7554/elife.96755] [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: 11/08/2024] Open
Abstract
Axonemal protein complexes, including the outer and inner dynein arms (ODA/IDA), are highly ordered structures of the sperm flagella that drive sperm motility. Deficiencies in several axonemal proteins have been associated with male infertility, which is characterized by asthenozoospermia or asthenoteratozoospermia. Dynein axonemal heavy chain 3 (DNAH3) resides in the IDA and is highly expressed in the testis. However, the relationship between DNAH3 and male infertility is still unclear. Herein, we identified biallelic variants of DNAH3 in four unrelated Han Chinese infertile men with asthenoteratozoospermia through whole-exome sequencing (WES). These variants contributed to deficient DNAH3 expression in the patients' sperm flagella. Importantly, the patients represented the anomalous sperm flagellar morphology, and the flagellar ultrastructure was severely disrupted. Intriguingly, Dnah3 knockout (KO) male mice were also infertile, especially showing the severe reduction in sperm movement with the abnormal IDA and mitochondrion structure. Mechanically, nonfunctional DNAH3 expression resulted in decreased expression of IDA-associated proteins in the spermatozoa flagella of patients and KO mice, including DNAH1, DNAH6, and DNALI1, the deletion of which has been involved in disruption of sperm motility. Moreover, the infertility of patients with DNAH3 variants and Dnah3 KO mice could be rescued by intracytoplasmic sperm injection (ICSI) treatment. Our findings indicated that DNAH3 is a novel pathogenic gene for asthenoteratozoospermia and may further contribute to the diagnosis, genetic counseling, and prognosis of male infertility.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
- NHC Key Laboratory of Chronobiology, Sichuan UniversityChengduChina
| | - Gan Shen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Yihong Yang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan UniversityChengduChina
| | - Chuan Jiang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Tiechao Ruan
- Department of Pediatrics, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Xue Yang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Liangchai Zhuo
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Yingteng Zhang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Yangdi Ou
- West China School of Medicine, Sichuan UniversityChengduChina
| | - Xinya Zhao
- West China School of Basic Medicine and Forensic Medicine, Sichuan UniversityChengduChina
| | - Shunhua Long
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and ChildrenChongqingChina
| | - Xiangrong Tang
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and ChildrenChongqingChina
| | - Tingting Lin
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and ChildrenChongqingChina
| | - Ying Shen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan UniversityChengduChina
- NHC Key Laboratory of Chronobiology, Sichuan UniversityChengduChina
| |
Collapse
|
3
|
Li Y, Wang Y, Tan YQ, Yue Q, Guo Y, Yan R, Meng L, Zhai H, Tong L, Yuan Z, Li W, Wang C, Han S, Ren S, Yan Y, Wang W, Gao L, Tan C, Hu T, Zhang H, Liu L, Yang P, Jiang W, Ye Y, Tan H, Wang Y, Lu C, Li X, Xie J, Yuan G, Cui Y, Shen B, Wang C, Guan Y, Li W, Shi Q, Lin G, Ni T, Sun Z, Ye L, Vourekas A, Guo X, Lin M, Zheng K. The landscape of RNA binding proteins in mammalian spermatogenesis. Science 2024; 386:eadj8172. [PMID: 39208083 DOI: 10.1126/science.adj8172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Despite continuous expansion of the RNA binding protein (RBP) world, there is a lack of systematic understanding of RBPs in the mammalian testis, which harbors one of the most complex tissue transcriptomes. We adapted RNA interactome capture to mouse male germ cells, building an RBP atlas characterized by multiple layers of dynamics along spermatogenesis. Trapping of RNA-cross-linked peptides showed that the glutamic acid-arginine (ER) patch, a residue-coevolved polyampholytic element present in coiled coils, enhances RNA binding of its host RBPs. Deletion of this element in NONO (non-POU domain-containing octamer-binding protein) led to a defective mitosis-to-meiosis transition due to compromised NONO-RNA interactions. Whole-exome sequencing of over 1000 infertile men revealed a prominent role of RBPs in the human genetic architecture of male infertility and identified risk ER patch variants.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yuanyuan Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Qiuling Yue
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Andrology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University, Nanjing 210008, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ruoyu Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Huicong Zhai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lingxiu Tong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zihan Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wu Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Cuicui Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shenglin Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Sen Ren
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yitong Yan
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Weixu Wang
- Institute of Computational Biology, Helmholtz Center Munich, Munich 85764, Germany
| | - Lei Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chen Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Tongyao Hu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Hao Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Liya Liu
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Pinglan Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wanyin Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yiting Ye
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Huanhuan Tan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yanfeng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chenyu Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xin Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jie Xie
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Gege Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yiqiang Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Yichun Guan
- Center for Reproductive Medicine, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Qinghua Shi
- Division of Reproduction and Genetics, First Affiliated Hospital of USC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Zheng Sun
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Ye
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Anastasios Vourekas
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mingyan Lin
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
- Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213000, China
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, Fuzhou 350014, China
| | - Ke Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
4
|
Brattig-Correia R, Almeida JM, Wyrwoll MJ, Julca I, Sobral D, Misra CS, Di Persio S, Guilgur LG, Schuppe HC, Silva N, Prudêncio P, Nóvoa A, Leocádio AS, Bom J, Laurentino S, Mallo M, Kliesch S, Mutwil M, Rocha LM, Tüttelmann F, Becker JD, Navarro-Costa P. The conserved genetic program of male germ cells uncovers ancient regulators of human spermatogenesis. eLife 2024; 13:RP95774. [PMID: 39388236 PMCID: PMC11466473 DOI: 10.7554/elife.95774] [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: 10/12/2024] Open
Abstract
Male germ cells share a common origin across animal species, therefore they likely retain a conserved genetic program that defines their cellular identity. However, the unique evolutionary dynamics of male germ cells coupled with their widespread leaky transcription pose significant obstacles to the identification of the core spermatogenic program. Through network analysis of the spermatocyte transcriptome of vertebrate and invertebrate species, we describe the conserved evolutionary origin of metazoan male germ cells at the molecular level. We estimate the average functional requirement of a metazoan male germ cell to correspond to the expression of approximately 10,000 protein-coding genes, a third of which defines a genetic scaffold of deeply conserved genes that has been retained throughout evolution. Such scaffold contains a set of 79 functional associations between 104 gene expression regulators that represent a core component of the conserved genetic program of metazoan spermatogenesis. By genetically interfering with the acquisition and maintenance of male germ cell identity, we uncover 161 previously unknown spermatogenesis genes and three new potential genetic causes of human infertility. These findings emphasize the importance of evolutionary history on human reproductive disease and establish a cross-species analytical pipeline that can be repurposed to other cell types and pathologies.
Collapse
Affiliation(s)
- Rion Brattig-Correia
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Department of Systems Science and Industrial Engineering, Binghamton UniversityNew YorkUnited States
| | - Joana M Almeida
- Instituto Gulbenkian de CiênciaOeirasPortugal
- EvoReproMed Lab, Environmental Health Institute (ISAMB), Associate Laboratory TERRA, Faculty of Medicine, University of LisbonLisbonPortugal
| | - Margot Julia Wyrwoll
- Centre of Medical Genetics, Institute of Reproductive Genetics, University and University Hospital of MünsterMünsterGermany
| | - Irene Julca
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Daniel Sobral
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University LisbonLisbonPortugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University LisbonCaparicaPortugal
| | - Chandra Shekhar Misra
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de LisboaOeirasPortugal
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | | | - Hans-Christian Schuppe
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig-UniversityGiessenGermany
| | - Neide Silva
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Pedro Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de LisboaLisboaPortugal
| | - Ana Nóvoa
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | | | - Joana Bom
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Sandra Laurentino
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | | | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital MünsterMünsterGermany
| | - Marek Mutwil
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Luis M Rocha
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Department of Systems Science and Industrial Engineering, Binghamton UniversityNew YorkUnited States
| | - Frank Tüttelmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University and University Hospital of MünsterMünsterGermany
| | - Jörg D Becker
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de LisboaOeirasPortugal
| | - Paulo Navarro-Costa
- Instituto Gulbenkian de CiênciaOeirasPortugal
- EvoReproMed Lab, Environmental Health Institute (ISAMB), Associate Laboratory TERRA, Faculty of Medicine, University of LisbonLisbonPortugal
| |
Collapse
|
5
|
Sethi S, Mehta P, Andrabi W, Mitra K, Rajender S. SPEM1 Gene Mutation in a Case with Sperm Morphological Defects Leading to Male Infertility. Reprod Sci 2024; 31:3102-3111. [PMID: 38886283 DOI: 10.1007/s43032-024-01612-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: 12/21/2023] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
The present study aimed at identifying the genetic mutation responsible for teratozoospermic infertility in a case with coiled sperm tails. A 33-year-old infertile male was diagnosed with teratozoospermic infertility, with sperm head in coiled (HIC) tail as the most common deformity. We employed whole exome sequencing to identify the genetic cause in this case. Exome sequencing data was filtered using the following criteria: MAF (< 0.003), ALFA project (< 0.001), 1000 Genomes (< 0.003), Granthem (> 50), Polyphen-2 (> 0.70), SIFT (< 0.03), and PhyloP (> = 0) scores. Shortlisted variants were looked in the in-house 29 exomes data available with us, and the variants that affected conserved amino acid residues or led to insertion/deletion or to protein-truncation with a Combined Annotation Dependent Depletion (CADD) score ≥ 10 were shortlisted. The variants thus populated were prioritized according to their roles in spermiogenesis. The study identified a heterozygous mutation c.826C > T (Arg276Trp) in the SPEM1 gene as a potential pathogenic variant that led to teratozoospermic infertility in the case under investigation. The mutation had a minor allele frequency of 0.00008176 in the gnomAd database and was absent in the Indian Genome Variations database. This is the first human study reporting a mutation in the SPEM1 gene as a cause of coiled sperm tails.
Collapse
Affiliation(s)
- Shruti Sethi
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Poonam Mehta
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | | | - Kalyan Mitra
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Singh Rajender
- Central Drug Research Institute, Lucknow, Uttar Pradesh, India.
- Academy of Scientific and Industrial Research (AcSIR), Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
6
|
Garcia TX, Matzuk MM. Novel Genes of the Male Reproductive System: Potential Roles in Male Reproduction and as Non-hormonal Male Contraceptive Targets. Mol Reprod Dev 2024; 91:e70000. [PMID: 39422082 DOI: 10.1002/mrd.70000] [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: 06/11/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
The development of novel non-hormonal male contraceptives represents a pivotal frontier in reproductive health, driven by the need for safe, effective, and reversible contraceptive methods. This comprehensive review explores the genetic underpinnings of male fertility, emphasizing the crucial roles of specific genes and structural variants (SVs) identified through advanced sequencing technologies such as long-read sequencing (LRS). LRS has revolutionized the detection of structural variants and complex genomic regions, offering unprecedented precision and resolution over traditional next-generation sequencing (NGS). Key genetic targets, including those implicated in spermatogenesis and sperm motility, are highlighted, showcasing their potential as non-hormonal contraceptive targets. The review delves into the systematic identification and validation of male reproductive tract-specific genes, utilizing advanced transcriptomics and genomics studies with validation using novel knockout mouse models. We discuss the innovative application of small molecule inhibitors, developed through platforms like DNA-encoded chemistry technology (DEC-Tec), which have shown significant promise in preclinical models. Notable examples include inhibitors targeting serine/threonine kinase 33 (STK33), soluble adenylyl cyclase (sAC), cyclin-dependent kinase 2 (CDK2), and bromodomain testis associated (BRDT), each demonstrating nanomolar affinity and potential for reversible and specific inhibition of male fertility. This review also honors the contributions of Dr. David L. Garbers whose foundational work has paved the way for these advancements. The integration of genomic, proteomic, and chemical biology approaches, supported by interdisciplinary collaboration, is poised to transform male contraceptive development. Future perspectives emphasize the need for continued innovation and rigorous testing to bring these novel contraceptives from the laboratory to clinical application, promising a new era of male reproductive health management.
Collapse
Affiliation(s)
- Thomas X Garcia
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Scott Department of Urology, Baylor College of Medicine, Houston, Texas, USA
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
Dinh TH, Phuong Anh N, Thao DH, Duy LD, Bac ND, Quyet PV, Son TT, Lan Anh LT, Canh NX, Hai NV, Duong NT. Single nucleotide polymorphisms of CFAP43 and TEX14 associated with idiopathic male infertility in a Vietnamese population. Medicine (Baltimore) 2024; 103:e39839. [PMID: 39331878 PMCID: PMC11441965 DOI: 10.1097/md.0000000000039839] [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: 05/19/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024] Open
Abstract
Male infertility is a multifactorial disease due to spermatogenesis impairment, with etiology remaining unknown for roughly one-third of infertile cases. Several studies have demonstrated that genetic variants are male infertility risk factors. CFAP43 and TEX14 are involved in the spermatogenesis process. The present study aimed to assess the association between single-nucleotide polymorphisms (SNPs) in CFAP43 (rs17116635 and rs10883979) and TEX14 (rs79813370 and rs34818467) and idiopathic male infertility in a Vietnamese population. A cohort of 206 infertile men and 195 controls were recruited for the study. CFAP43 and TEX14 SNPs were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Genotypes of randomly selected samples, accounting for 10% of the total, were confirmed using Sanger sequencing. The obtained data were analyzed using statistical methods. The results showed that 4 SNPs (rs17116635, rs10883979, rs79813370, and rs34818467) were in accordance with Hardy-Weinberg Equilibrium (HWE; P > .05). CFAP43 rs10883979 and TEX14 rs79813370 were associated with male infertility. For CFAP43 rs10883979, in the recessive model, the combination AA + AG was associated with male infertility when compared to the GG genotype (OR = 0.26; 95% CI: 0.06-0.85; P = .02). For TEX14 rs79813370, a protective effect against infertility risk was identified in the presence of the T allele of rs79813370 when compared to the G allele (OR = 0.48; 95% CI: 0.32-0.72; P < .001). Our results suggest that CFAP43 rs10883979 and TEX14 rs79813370 are likely associated with male infertility in the Vietnamese population, in which the G allele of rs79813370 may be a risk factor for male infertility.
Collapse
Affiliation(s)
- Tran Huu Dinh
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Nguyen Phuong Anh
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Dinh Huong Thao
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - La Duc Duy
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Nguyen Duy Bac
- Department of Human Anatomy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Pham Van Quyet
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
- Military Institute of Clinical Embryology and Histology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Trinh The Son
- Military Institute of Clinical Embryology and Histology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Luong Thi Lan Anh
- Department of Medical Biology and Genetics, Hanoi Medical University, Ministry of Health, Hanoi, Vietnam
| | - Nguyen Xuan Canh
- Department of Microbial Biotechnology, Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Nong Van Hai
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Nguyen Thuy Duong
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| |
Collapse
|
8
|
Ogloblinsky MSC, Bocher O, Aloui C, Leutenegger AL, Ozisik O, Baudot A, Tournier-Lasserve E, Castillo-Madeen H, Lewinsohn D, Conrad DF, Génin E, Marenne G. PSAP-Genomic-Regions: A Method Leveraging Population Data to Prioritize Coding and Non-Coding Variants in Whole Genome Sequencing for Rare Disease Diagnosis. Genet Epidemiol 2024. [PMID: 39318036 DOI: 10.1002/gepi.22593] [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: 06/05/2024] [Revised: 07/30/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
The introduction of Next-Generation Sequencing technologies in the clinics has improved rare disease diagnosis. Nonetheless, for very heterogeneous or very rare diseases, more than half of cases still lack molecular diagnosis. Novel strategies are needed to prioritize variants within a single individual. The Population Sampling Probability (PSAP) method was developed to meet this aim but only for coding variants in exome data. Here, we propose an extension of the PSAP method to the non-coding genome called PSAP-genomic-regions. In this extension, instead of considering genes as testing units (PSAP-genes strategy), we use genomic regions defined over the whole genome that pinpoint potential functional constraints. We conceived an evaluation protocol for our method using artificially generated disease exomes and genomes, by inserting coding and non-coding pathogenic ClinVar variants in large data sets of exomes and genomes from the general population. PSAP-genomic-regions significantly improves the ranking of these variants compared to using a pathogenicity score alone. Using PSAP-genomic-regions, more than 50% of non-coding ClinVar variants were among the top 10 variants of the genome. On real sequencing data from six patients with Cerebral Small Vessel Disease and nine patients with male infertility, all causal variants were ranked in the top 100 variants with PSAP-genomic-regions. By revisiting the testing units used in the PSAP method to include non-coding variants, we have developed PSAP-genomic-regions, an efficient whole-genome prioritization tool which offers promising results for the diagnosis of unresolved rare diseases.
Collapse
Affiliation(s)
| | - Ozvan Bocher
- Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- Institute of Translational Genomics, Helmholtz Zentrum München, Munich, Germany
| | - Chaker Aloui
- Inserm, NeuroDiderot, Unité Mixte de Recherche, Université Paris Cité, Paris, France
| | | | - Ozan Ozisik
- INSERM, Marseille Medical Genetics (MMG), Aix Marseille University, Marseille, France
| | - Anaïs Baudot
- INSERM, Marseille Medical Genetics (MMG), Aix Marseille University, Marseille, France
| | - Elisabeth Tournier-Lasserve
- Inserm, NeuroDiderot, Unité Mixte de Recherche, Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Service de Génétique Moléculaire Neurovasculaire, Hôpital Saint-Louis, Paris, France
| | - Helen Castillo-Madeen
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Daniel Lewinsohn
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Emmanuelle Génin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- Centre Hospitalier Régional Universitaire de Brest, Brest, France
| | | |
Collapse
|
9
|
Yi Zhou B, Ting Fu W, Gu H, Zhen Li M, Bin Zhong X, Tang J. A retrospective analysis of 1600 infertility patients with azoospermia and severe oligozoospermia. Clin Chim Acta 2024; 565:119973. [PMID: 39307333 DOI: 10.1016/j.cca.2024.119973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE This study aimed to investigate the genetic etiology of male infertility patients. METHOD A total of 1600 male patients with infertility, including 1300 cases of azoospermia and 300 cases of severe oligozoospermia, underwent routine semen analysis, chromosomal karyotype analysis and sex hormone level testing. The Azoospermia factor (AZF) on the Y chromosome was detected using the multiple fluorescence quantitative PCR technique. Additionally, copy number variation (CNV) analysis was performed on patients with Sertoli-cell-only syndrome who had a normal karyotype and AZF. RESULT Chromosomal abnormalities were found in 334 cases (20.88 %) of the 1600 male infertility patients. The most common type of abnormality was sex chromosome abnormalities (18.94 %), with 47, XXY being the most frequent abnormal karyotype. The rates of chromosomal abnormalities were significantly different between the azoospermia group and the severe oligospermia group (23.69 % and 8.67 %, respectively; P<0.05). AZF microdeletions were detected in 155 cases (9.69 %), with various deletion types and AZFc region microdeletion being the most prevalent. The rates of AZF microdeletions were not significantly different between the azoospermia group and the severe oligospermia group (9.15 % and 12 %, respectively; P=0.133). In 92 patients with Sertoli-cell-only syndrome who had a normal karyotype and AZF, the detection rate of CNV was 16.3 %. Compared to the severe oligospermia group, the azoospermia group had higher levels of FSH and LH and lower levels of T and E2, and the differences were statistically significant (P<0.05). CONCLUSIONS Male infertility is a complex multifactorial disease, with chromosomal abnormalities and Y chromosome microdeletions being important genetic factors leading to the disease. Initial genetic testing of infertile men should include karyotyping and Y chromosome microdeletions. If necessary, CNV testing should be performed to establish a clinical diagnosis and provide individualized treatment for male infertility.
Collapse
Affiliation(s)
- Bing Yi Zhou
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510060, China
| | - Wen Ting Fu
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510060, China
| | - Heng Gu
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510060, China
| | - Ming Zhen Li
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510060, China
| | - Xiao Bin Zhong
- School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jia Tang
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou 510060, China; School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
10
|
Riera-Escamilla A, Nagirnaja L. Utility of exome sequencing in primary spermatogenic disorders: From research to diagnostics. Andrology 2024. [PMID: 39300832 DOI: 10.1111/andr.13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Primary spermatogenic disorders represent a severe form of male infertility whereby sperm production is impaired due to testicular dysfunction, leading to reduced quality or quantity of spermatozoa. Gene-centered research has certainly demonstrated the importance of the genetic factor in the etiology of both poor sperm morphology or motility and reduced sperm count. In the last decade, next-generation sequencing has expanded the research to whole exome which has transformed our understanding of male infertility genetics, but uncertainty persists in its diagnostic yield, especially in large unrelated populations. OBJECTIVE To evaluate the utility of exome sequencing in detecting genetic factors contributing to various traits of primary spermatogenic disorders, which is a crucial step before interpreting the diagnostic yield of the platform. MATERIALS AND METHODS We manually curated 415 manuscripts and included 19 research studies that predominantly performed whole exome sequencing in cohorts of unrelated cases with primary spermatogenic defects. RESULTS The detection rate, defined as the fraction of cases with an identifiable genetic cause, typically remained below 25% for quantitative defects of spermatozoa, whereas improved rates were observed for traits of abnormal sperm morphology/motility and in populations enriched with consanguineous families. Unlike the quantitative defects, the genetic architecture of the qualitative issues of spermatozoa featured a small number of recurrent genes describing a large fraction of studied cases. These observations were also in line with the lower biological complexity of the pathways affected by the reported genes. DISCUSSION AND CONCLUSIONS This review demonstrates the variability in detection rates of exome sequencing across semen phenotypes, which may have an impact on the expectations of the diagnostic yield in the clinical setting.
Collapse
Affiliation(s)
- Antoni Riera-Escamilla
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
11
|
Motawi A, Crafa A, Hamoda T, Shah R, Agarwal A. The Andrological Landscape in the Twenty-First Century: Making Sense of the Advances in Male Infertility Management for the Busy Clinicians. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1222. [PMID: 39338105 PMCID: PMC11431684 DOI: 10.3390/ijerph21091222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Male infertility represents a significant global problem due to its essential health, social, and economic implications. It is unsurprising that scientific research is very active in this area and that advances in the diagnostic and therapeutic fields are notable. This review presents the main diagnostic advances in male infertility, starting from the changes made in the latest WHO Manual of semen analysis and discussing the more molecular aspects inherent to "omics". Furthermore, the usefulness of artificial intelligence in male infertility diagnostics and the latest advances in varicocele diagnosis will be discussed. In particular, the diagnostic path of male infertility is increasingly moving towards a personalized approach to the search for the specific biomarkers of infertility and the prediction of treatment response. The treatment of male infertility remains empirical in many regards, but despite that, advances have been made to help formulate evidence-based recommendations. Varicocele, the most common correctable cause of male infertility, has been explored for expanded indications for repair. The following expanded indications were discussed: elevated sperm DNA fragmentation, hypogonadism, orchalgia, and the role of varicocele repair in non-obstructive azoospermia. Moving forward with the available data, we discussed the stepwise approach to surgical sperm retrieval techniques and the current measures that have been investigated for optimizing such patients before testicular sperm extraction. Finally, the key points and expert recommendations regarding the best practice for diagnosing and treating men with infertility were summarized to conclude this review.
Collapse
Affiliation(s)
- Ahmad Motawi
- Department of Andrology, Sexual Medicine and STIs, Faculty of Medicine, Cairo University, Cairo 11956, Egypt
- Global Andrology Forum (GAF), Moreland Hills, OH 44022, USA
| | - Andrea Crafa
- Global Andrology Forum (GAF), Moreland Hills, OH 44022, USA
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Taha Hamoda
- Global Andrology Forum (GAF), Moreland Hills, OH 44022, USA
- Department of Urology, King Abdulaziz University, Jeddah P.O. Box 80215, Saudi Arabia
- Department of Urology, Faculty of Medicine, Minia University, Minia 2431436, Egypt
| | - Rupin Shah
- Global Andrology Forum (GAF), Moreland Hills, OH 44022, USA
- Department of Urology, Lilavati Hospital and Research Center, Mumbai 400050, India
| | - Ashok Agarwal
- Global Andrology Forum (GAF), Moreland Hills, OH 44022, USA
| |
Collapse
|
12
|
Veltman JA, Tüttelmann F. Why geneticists should care about male infertility. Nat Rev Genet 2024:10.1038/s41576-024-00773-3. [PMID: 39294342 DOI: 10.1038/s41576-024-00773-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Affiliation(s)
- Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Frank Tüttelmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Münster, Germany
| |
Collapse
|
13
|
Zhou H, Yin Z, Ni B, Lin J, Luo S, Xie W. Whole exome sequencing analysis of 167 men with primary infertility. BMC Med Genomics 2024; 17:230. [PMID: 39267058 PMCID: PMC11391607 DOI: 10.1186/s12920-024-02005-3] [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: 06/28/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Spermatogenic failure is one of the leading causes of male infertility and its genetic etiology has not yet been fully understood. METHODS The study screened a cohort of patients (n = 167) with primary male infertility in contrast to 210 normally fertile men using whole exome sequencing (WES). The expression analysis of the candidate genes based on public single cell sequencing data was performed using the R language Seurat package. RESULTS No pathogenic copy number variations (CNVs) related to male infertility were identified using the the GATK-gCNV tool. Accordingly, variants of 17 known causative (five X-linked and twelve autosomal) genes, including ACTRT1, ADAD2, AR, BCORL1, CFAP47, CFAP54, DNAH17, DNAH6, DNAH7, DNAH8, DNAH9, FSIP2, MSH4, SLC9C1, TDRD9, TTC21A, and WNK3, were identified in 23 patients. Variants of 12 candidate (seven X-linked and five autosomal) genes were identified, among which CHTF18, DDB1, DNAH12, FANCB, GALNT3, OPHN1, SCML2, UPF3A, and ZMYM3 had altered fertility and semen characteristics in previously described knockout mouse models, whereas MAGEC1,RBMXL3, and ZNF185 were recurrently detected in patients with male factor infertility. The human testis single cell-sequencing database reveals that CHTF18, DDB1 and MAGEC1 are preferentially expressed in spermatogonial stem cells. DNAH12 and GALNT3 are found primarily in spermatocytes and early spermatids. UPF3A is present at a high level throughout spermatogenesis except in elongating spermatids. The testicular expression profiles of these candidate genes underlie their potential roles in spermatogenesis and the pathogenesis of male infertility. CONCLUSION WES is an effective tool in the genetic diagnosis of primary male infertility. Our findings provide useful information on precise treatment, genetic counseling, and birth defect prevention for male factor infertility.
Collapse
Affiliation(s)
- Haiyan Zhou
- National Health Commission Key Laboratory of Birth Defects for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Zhaochu Yin
- National Health Commission Key Laboratory of Birth Defects for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Bin Ni
- National Health Commission Key Laboratory of Birth Defects for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Jiwu Lin
- National Health Commission Key Laboratory of Birth Defects for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Shuwei Luo
- Center for Reproductive Medicine, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China.
- Hunan Provincial Maternal and Child Health Care Hospital, No. 53 Xiangchun Road, Changsha, Hunan, 410008, China.
| | - Wanqin Xie
- National Health Commission Key Laboratory of Birth Defects for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China.
- Hunan Provincial Maternal and Child Health Care Hospital, No. 53 Xiangchun Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
14
|
Zemet R, Van den Veyver IB. Impact of prenatal genomics on clinical genetics practice. Best Pract Res Clin Obstet Gynaecol 2024; 97:102545. [PMID: 39265228 DOI: 10.1016/j.bpobgyn.2024.102545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/18/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Genetic testing for prenatal diagnosis in the pre-genomic era primarily focused on detecting common fetal aneuploidies, using methods that combine maternal factors and imaging findings. The genomic era, ushered in by the emergence of new technologies like chromosomal microarray analysis and next-generation sequencing, has transformed prenatal diagnosis. These new tools enable screening and testing for a broad spectrum of genetic conditions, from chromosomal to monogenic disorders, and significantly enhance diagnostic precision and efficacy. This chapter reviews the transition from traditional karyotyping to comprehensive sequencing-based genomic analyses. We discuss both the clinical utility and the challenges of integrating prenatal exome and genome sequencing into prenatal care and underscore the need for ethical frameworks, improved prenatal phenotypic characterization, and global collaboration to further advance the field.
Collapse
Affiliation(s)
- Roni Zemet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - Ignatia B Van den Veyver
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Division of Prenatal and Reproductive Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Chen D, Fan G, Xu Y, Luo P, Chen Q, Chen X, Guo Z, Zhu X, Gao Y. A novel homozygous mutation in the DNAAF3 gene leads to severe asthenozoospermia and teratospermia. J Cell Mol Med 2024; 28:e70092. [PMID: 39289782 PMCID: PMC11408122 DOI: 10.1111/jcmm.70092] [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: 11/22/2023] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Primary ciliary dyskinesia (PCD) is an autosomal recessive genetic disorder characterized by ultrastructural defects in the cilia or flagella of cells, causing respiratory abnormalities, sinusitis, visceral transposition, and male infertility. DNAAF3 plays an important role in the assembly and transportation of axonemal dynein complexes in cilia or flagella and has been shown to be associated with PCD. To date, only two cases of PCD with infertility associated with DNAAF3 mutations have been reported, and no mouse models for this gene have been successfully constructed. This study was conducted on an infertile Chinese male patient with a history of bronchitis. Examination of the patient's semen revealed severe asthenozoospermia and teratospermia. Whole exome sequencing revealed a new homozygous loss-of-function DNAAF3 mutation. CRISPR-Cas9 gene-editing technology was used to construct the same mutation in C57/B6 mice, revealing that homozygous C57/B6 mice were characterized by severe hydrocephalus and early death. The results of this study expand the mutation spectrum of DNAAF3 and confirm its correlation with PCD pathogenesis. This study provides new insights on the mechanisms underlying male infertility related to DNAAF3 mutation and PCD.
Collapse
Affiliation(s)
- Dongjia Chen
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Guoqing Fan
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yan Xu
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Peng Luo
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Qinyun Chen
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xuren Chen
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zexin Guo
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xianqing Zhu
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yong Gao
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
16
|
Shi F, Liu Y, Chen Z, Li D, Yao Y, Zhou M, Zhuo Y, Ma X, Cao D. An integrated approach for improving clinical management of non-obstructive azoospermia. Andrology 2024; 12:1312-1323. [PMID: 38221731 DOI: 10.1111/andr.13587] [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: 05/10/2023] [Revised: 11/06/2023] [Accepted: 12/19/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Non-obstructive azoospermia is the most severe form of male infertility. A testicular biopsy is required for the diagnosis of non-obstructive azoospermia, and the causal factors for non-obstructive azoospermia remain unknown. OBJECTIVES To reduce the risk of multiple biopsies and identify factors that contribute to non-obstructive azoospermia, we proposed an integrated approach for the preoperative diagnosis and clinical management of non-obstructive azoospermia by applying the chromosome-spreading technique and whole-exome sequencing. MATERIALS AND METHODS Between July 2020 and December 2022, after ruling out definitive obstructive azoospermia and non-obstructive azoospermia patients with testicular volume < 6 mL, 20 patients with non-obstructive azoospermia who underwent preoperative testicular diagnostic biopsy using testicular sperm aspiration were subjected to retrospective analysis. RESULTS Microscopic examination identified four patients with sperm cells, and 16 without sperm cells. Routine pathological analysis classified one patient as normal spermatogenesis, three as hypospermatogenesis, five as maturation arrest, nine as Sertoli cell-only, and two as unable to judge. With chromosome-spreading technology using routine cell suspension samples for microscopic examination, 18 patient diagnoses were validated, and two patients without a definitive diagnosis were supplemented. Detection of the Y chromosome and a well-organized whole-exome sequencing analysis revealed potential genetic factors. DISCUSSION AND CONCLUSION The full use of testicular biopsy is beneficial for the diagnosis of azoospermia, as it avoids the risk of multiple biopsies. Moreover, in combination with whole-exome sequencing, clinicians can obtain more information regarding the pathogenesis of non-obstructive azoospermia, which may guide treatment.
Collapse
Affiliation(s)
- Fu Shi
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Ye Liu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Zheng Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongliang Li
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Meixun Zhou
- Department of Pathology, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Yumin Zhuo
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xin Ma
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Senior Department of Urology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Dandan Cao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
17
|
Oud MS, de Leeuw N, Smeets DFCM, Ramos L, van der Heijden GW, Timmermans RGJ, van de Vorst M, Hofste T, Kempers MJE, Stokman MF, D'Hauwers KWM, Faas BHW, Westra D. Innovative all-in-one exome sequencing strategy for diagnostic genetic testing in male infertility: Validation and 10-month experience. Andrology 2024. [PMID: 39180390 DOI: 10.1111/andr.13742] [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: 04/12/2024] [Revised: 07/16/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Current guidelines indicate that patients with extreme oligozoospermia or azoospermia should be tested for chromosomal imbalances, azoospermia factor (AZF) deletions and/or CFTR variants. For other sperm abnormalities, no genetic diagnostics are recommended. OBJECTIVES To determine whether exome sequencing (ES) with combined copy number variant (CNV) and single nucleotide variant (SNV) analysis is a reliable first-tier method to replace current methods (validation study), and to evaluate the diagnostic yield after 10 months of implementation (evaluation study). MATERIALS AND METHODS In the validation study, ES was performed on DNA of patients already diagnosed with AZF deletions (n = 17), (non-)mosaic sex chromosomal aneuploidies or structural chromosomal anomalies (n = 37), CFTR variants (n = 26), or variants in known infertility genes (n = 4), and 90 controls. The data were analyzed using our standard diagnostic pipeline, with a bioinformatic filter for 130 male infertility genes. In the evaluation study, results of 292 clinical exomes were included. RESULTS All previously reported variants in the validation cohort, including clinically relevant Y-chromosomal microdeletions, were correctly identified and reliably detected. In the evaluation study, we identified one or more clinically relevant genetic anomalies in 67 of 292 of all cases (22.9%): these included aberrations that could have been detected with current methods in 30 of 67 patients (10.2% of total), (possible) (mono)genetic causes in the male infertility gene panel in 28 of 67 patients (9.6%), and carriership of cystic fibrosis in nine of 67 patients (3.1%). CONCLUSION ES is a reliable first-tier method to detect the most common genetic causes of male infertility and, as additional genetic causes can be detected, in our evaluation cohort the diagnostic yield almost doubled (10.2%-19.8%, excluding CF carriers). A genetic diagnosis provides answers on the cause of infertility and helps the professionals in the counseling for treatment, possible co-morbidities and risk for offspring and/or family members. Karyotyping will still remain necessary for detecting balanced translocations or low-grade chromosomal mosaicism.
Collapse
Affiliation(s)
- Manon S Oud
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Nicole de Leeuw
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Dominique F C M Smeets
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Liliana Ramos
- Department of Obstetrics and Gynaecology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Raoul G J Timmermans
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Maartje van de Vorst
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Tom Hofste
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Marlies J E Kempers
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Marijn F Stokman
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Brigitte H W Faas
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Dineke Westra
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Stallmeyer B, Dicke AK, Tüttelmann F. How exome sequencing improves the diagnostics and management of men with non-syndromic infertility. Andrology 2024. [PMID: 39120565 DOI: 10.1111/andr.13728] [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: 03/03/2024] [Revised: 07/07/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
Male infertility affects approximately 17% of all men and represents a complex disorder in which not only semen parameters such as sperm motility, morphology, and number of sperm are highly variable, but also testicular phenotypes range from normal spermatogenesis to complete absence of germ cells. Genetic factors significantly contribute to the disease but chromosomal aberrations, mostly Klinefelter syndrome, and microdeletions of the Y-chromosome have remained the only diagnostically and clinically considered genetic causes. Monogenic causes remain understudied and, thus, often unidentified, leaving the majority of the male factor couple infertility pathomechanistically unexplained. This has been changing mostly because of the introduction of exome sequencing that allows the analysis of multiple genes in large patient cohorts. As a result, pathogenic variants in single genes have been associated with non-syndromic forms of all aetiologic sub-categories in the last decade. This review highlights the contribution of exome sequencing to the identification of novel disease genes for isolated (non-syndromic) male infertility by presenting the results of a comprehensive literature search. Both, reduced sperm count in azoospermic and oligozoospermic patients, and impaired sperm motility and/or morphology, in asthenozoospermic and/or teratozoospermic patients are highly heterogeneous diseases with well over 100 different candidate genes described for each entity. Applying the standardized evaluation criteria of the ClinGen gene curation working group, 70 genes with at least moderate evidence to contribute to the disease are highlighted. The implementation of these valid disease genes in clinical exome sequencing is important to increase the diagnostic yield in male infertility and, thus, improve clinical decision-making and appropriate genetic counseling. Future advances in androgenetics will continue to depend on large-scale exome and genome sequencing studies of comprehensive international patient cohorts, which are the most promising approaches to identify additional disease genes and provide reliable data on the gene-disease relationship.
Collapse
Affiliation(s)
- Birgit Stallmeyer
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Munster, Germany
| | - Ann-Kristin Dicke
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Munster, Germany
| | - Frank Tüttelmann
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Munster, Germany
| |
Collapse
|
19
|
Okutman Ö, Gürbüz AS, Salvarci A, Büyük U, Ruso H, Gürgan T, Tarabeux J, Leuvrey AS, Nourisson E, Lang C, Muller J, Viville S. Evaluation of an Updated Gene Panel as a Diagnostic Tool for Both Male and Female Infertility. Reprod Sci 2024; 31:2309-2317. [PMID: 38664359 DOI: 10.1007/s43032-024-01553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/02/2024] [Indexed: 07/31/2024]
Abstract
In recent years, an increasing number of genes associated with male and female infertility have been identified. The genetics of infertility is no longer limited to the analysis of karyotypes or specific genes, and it is now possible to analyse several dozen infertility genes simultaneously. Here, we present the diagnostic activity over the past two years including 140 patients (63 women and 77 men). Targeted sequencing revealed causative variants in 17 patients, representing an overall diagnostic rate of 12.1%, with prevalence rates in females and males of 11% and 13%, respectively. The gene-disease relationship (GDR) was re-evaluated for genes due to the addition of new patients and/or variants in the actual study. Five genes changed categories: two female genes (MEIOB and TBPL2) moved from limited to moderate; two male genes (SOHLH1 and GALNTL5) moved from no evidence to strong and from limited to moderate; and SEPTIN12, which was unable to classify male infertility, was reclassified as limited. Many infertility genes have yet to be identified. With the increasing integration of genetics in reproductive medicine, the scope of intervention extends to include other family members, in addition to individual patients or couples. Genetic counselling consultations and appropriate staffing will need to be established in fertility centres. Trial registration number: Not applicable.
Collapse
Affiliation(s)
- Özlem Okutman
- Service de Gynécologie-Obstetrique, Clinique de Fertilité, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Hôpital Erasme, Route de Lennik, 808, 1070, Brussels, Belgium.
| | | | | | - Umut Büyük
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkey
| | - Halil Ruso
- Gürgan Clinic Women's Health and IVF Centre, Ankara, Turkey
- Faculty of Medicine, Department of Histology and Embryology, Gazi University, Ankara, Turkey
| | - Timur Gürgan
- Gürgan Clinic Women's Health and IVF Centre, Ankara, Turkey
- Department of Obstetrics and Gynecology, Bahçeşehir University School of Medicine, Istanbul, Turkey
| | - Julien Tarabeux
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Anne-Sophie Leuvrey
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Elsa Nourisson
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Cécile Lang
- Laboratoire de Diagnostic Génétique, Unité de Génétique de L'infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean Muller
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d'Alsace (IGMA), Université de Strasbourg, INSERM UMR 1112, Strasbourg, France
- Unité Fonctionnelle de Bioinformatique Médicale Appliquée Au Diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Stephane Viville
- Laboratoire de Diagnostic Génétique, Unité de Génétique de L'infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d'Alsace (IGMA), Université de Strasbourg, INSERM UMR 1112, Strasbourg, France
| |
Collapse
|
20
|
Wu H, Zhang Y, Li Y, Sun S, Zhang J, Xie Q, Dong Y, Zhou S, Sha X, Li K, Chen J, Zhang X, Gao Y, Shen Q, Wang G, Zha X, Duan Z, Tang D, Xu C, Geng H, Lv M, Xu Y, Zhou P, Wei Z, Hua R, Cao Y, Liu M, He X. Adenylate kinase phosphate energy shuttle underlies energetic communication in flagellar axonemes. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1697-1714. [PMID: 38761355 DOI: 10.1007/s11427-023-2539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/29/2024] [Indexed: 05/20/2024]
Abstract
The complexities of energy transfer mechanisms in the flagella of mammalian sperm flagella have been intensively investigated and demonstrate significant diversity across species. Enzymatic shuttles, particularly adenylate kinase (AK) and creatine kinase (CK), are pivotal in the efficient transfer of intracellular ATP, showing distinct tissue- and species-specificity. Here, the expression profiles of AK and CK were investigated in mice and found to fall into four subgroups, of which Subgroup III AKs were observed to be unique to the male reproductive system and conserved across chordates. Both AK8 and AK9 were found to be indispensable to male reproduction after analysis of an infertile male cohort. Knockout mouse models showed that AK8 and AK9 were central to promoting sperm motility. Immunoprecipitation combined with mass spectrometry revealed that AK8 and AK9 interact with the radial spoke (RS) of the axoneme. Examination of various human and mouse sperm samples with substructural damage, including the presence of multiple RS subunits, showed that the head of radial spoke 3 acts as an adapter for AK9 in the flagellar axoneme. Using an ATP probe together with metabolomic analysis, it was found that AK8 and AK9 cooperatively regulated ATP transfer in the axoneme, and were concentrated at sites associated with energy consumption in the flagellum. These findings indicate a novel function for RS beyond its structural role, namely, the regulation of ATP transfer. In conclusion, the results expand the functional spectrum of AK proteins and suggest a fresh model regarding ATP transfer within mammalian flagella.
Collapse
Affiliation(s)
- Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Yanman Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Yuqian Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shuya Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Jintao Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qingsong Xie
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yue Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Shushu Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Xuan Sha
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Kuokuo Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Jinyi Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Qunshan Shen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Guanxiong Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaomin Zha
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Zongliu Duan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Chuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Yuping Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, China
| | - Rong Hua
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China.
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, China.
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, China.
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
21
|
Pozzi E, Belladelli F, Corsini C, Boeri L, Capogrosso P, Fallara G, Candela L, Bertini A, Cattafi F, Raffo M, Cazzaniga W, Ventimiglia E, d'Arma A, Alfano M, Matloob R, Papaleo E, Candiani M, Montorsi F, Salonia A. Contemporary Diagnostic Work-Up for Male Infertility: Emphasizing Comprehensive Baseline Assessment. World J Mens Health 2024; 42:42.e63. [PMID: 39028127 DOI: 10.5534/wjmh.240069] [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: 03/18/2024] [Revised: 03/19/2024] [Accepted: 04/09/2024] [Indexed: 07/20/2024] Open
Abstract
Infertility is a prevalent issue affecting many couples during their reproductive years, with a significant number facing challenges in conceiving despite regular unprotected intercourse. Male factor infertility (MFI) contributes significantly to these cases, with a significant proportion of men lacking an identifiable etiology. As such, a thorough assessment of MFI has become increasingly vital for personalized management. This position paper from the Andrology team at IRCCS Ospedale San Raffaele emphasizes a comprehensive and individualized approach to MFI work-up, addressing the evolving challenges encountered in clinical practice. Our approach involves a thorough diagnostic work-up to identify the underlying causes of MFI, integrating insights from extensive literature review and our proprietary data. Our data demonstrates that an extensive diagnostic assessment allows us to identify at least one underlying cause of MFI in most infertile men. However, challenges persist in diagnosing less severe phenotypes with unclear etiology. We discuss the importance of individualized MFI work-up and its implications for developing rational therapeutic protocols. Lastly, this paper highlights the necessity for a personalized diagnostic assessment, addressing the daily clinical challenges and emphasizing tailored approaches to try to improve outcomes among couples seeking first medical help for infertility.
Collapse
Affiliation(s)
- Edoardo Pozzi
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federico Belladelli
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Christian Corsini
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Boeri
- Department of Urology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Capogrosso
- Department of Urology, Circolo & Fondazione Macchi Hospital - ASST Sette Laghi, Varese, Italy
| | - Giuseppe Fallara
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luigi Candela
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandro Bertini
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Cattafi
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimiliano Raffo
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Walter Cazzaniga
- Department of Urology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Eugenio Ventimiglia
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia d'Arma
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Rayan Matloob
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Enrico Papaleo
- Department of Obstetrics and Gynaecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Candiani
- University Vita-Salute San Raffaele, Milan, Italy
- Department of Obstetrics and Gynaecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- University Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
22
|
Navapour L, Mogharrab N, Parvin A, Rezaei Arablouydareh S, Movahedpour A, Jebraeily M, Taheri-Anganeh M, Ghasemnejad-Berenji H. Identification of high-risk non-synonymous SNPs (nsSNPs) in DNAH1 and DNAH17 genes associated with male infertility: a bioinformatics analysis. J Appl Genet 2024:10.1007/s13353-024-00884-x. [PMID: 38874855 DOI: 10.1007/s13353-024-00884-x] [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: 02/01/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
Male infertility is a significant reproductive issue affecting a considerable number of couples worldwide. While there are various causes of male infertility, genetic factors play a crucial role in its development. We focused on identifying and analyzing the high-risk nsSNPs in DNAH1 and DNAH17 genes, which encode proteins involved in sperm motility. A total of 20 nsSNPs for DNAH1 and 10 nsSNPs for DNAH17 were analyzed using various bioinformatics tools including SIFT, PolyPhen-2, CADD, PhD-SNPg, VEST-4, and MutPred2. As a result, V1287G, L2071R, R2356W, R3169C, R3229C, E3284K, R4096L, R4133C, and A4174T in DNAH1 gene and C1803Y, C1829Y, R1903C, and L3595P in DNAH17 gene were identified as high-risk nsSNPs. These nsSNPs were predicted to decrease protein stability, and almost all were found in highly conserved amino acid positions. Additionally, 4 nsSNPs were observed to alter post-translational modification status. Furthermore, the interaction network analysis revealed that DNAH1 and DNAH17 interact with DNAH2, DNAH3, DNAH5, DNAH7, DNAH8, DNAI2, DNAL1, CFAP70, DNAI3, DNAI4, ODAD1, and DNAI7, demonstrating the importance of DNAH1 and DNAH17 proteins in the overall functioning of the sperm motility machinery. Taken together, these findings revealed the detrimental effects of identified high-risk nsSNPs on protein structure and function and highlighted their potential relevance to male infertility. Further studies are warranted to validate these findings and to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Leila Navapour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Mogharrab
- Biophysics and Computational Biology Laboratory (BCBL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Ali Parvin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sahar Rezaei Arablouydareh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohamad Jebraeily
- Department of Health Information Technology, School of Allied Medical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
23
|
Zhang G, Ye F, Yang Y, Xiong D, Zhi W, Wu Y, Sun Y, Zeng J, Liu W. Identification of a novel mutation in chibby family member 2 in a non-obstructive azoospermic patient. Reprod Biol 2024; 24:100891. [PMID: 38733656 DOI: 10.1016/j.repbio.2024.100891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
Azoospermia constitutes a significant factor in male infertility, defined by the absence of spermatozoa in the ejaculate, afflicting 15% of infertile men. However, a subset of azoospermic cases remains unattributed to known genetic variants. Prior investigations have identified the chibby family member 2 (CBY2) as prominently and specifically expressed in the testes of both humans and mice, implicating its potential involvement in spermatogenesis. In this study, we conducted whole exome sequencing (WES) on an infertile family to uncover novel genetic factors contributing to azoospermia. Our analysis revealed a homozygous c .355 C>A variant of CBY2 in a non-obstructive azoospermic patient. This deleterious variant significantly diminished the protein expression of CBY2 both in vivo and in vitro, leading to a pronounced disruption of spermatogenesis at the early round spermatid stage post-meiosis. This disruption was characterized by a nearly complete loss of elongating and elongated spermatids. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) and co-immunoprecipitation assays demonstrated the interaction between CBY2 and Piwi-like protein 1 (PIWIL1). Immunofluorescence staining further confirmed the co-localization of CBY2 and PIWIL1 in the testes during the spermatogenic process in both humans and mice. Additionally, diminished PIWIL1 expression was observed in the testicular tissue from the affected patient. Our findings suggest that the homozygous c .355 C>A variant of CBY2 compromises CBY2 function, contributing to defective spermatogenesis at the round spermiogenic stage and implicating its role in the pathogenesis of azoospermia.
Collapse
Affiliation(s)
- Guohui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China; Reproductive Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China
| | - Fei Ye
- Reproductive Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China
| | - Yihong Yang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dongsheng Xiong
- Reproductive Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China
| | - Weiwei Zhi
- Reproductive Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China
| | - Yang Wu
- Reproductive Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610045, China
| | - Yongkang Sun
- Department of Obstetrics/Gynecology, 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
- Reproductive Medicine Center, 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
|
24
|
Barbotin AL, Boursier A, Jourdain AS, Moerman A, Rabat B, Chehimi M, Thuillier C, Ghoumid J, Smol T. Identification of a novel CFAP61 homozygous splicing variant associated with multiple morphological abnormalities of the flagella. J Assist Reprod Genet 2024; 41:1499-1505. [PMID: 38775994 PMCID: PMC11224159 DOI: 10.1007/s10815-024-03139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/07/2024] [Indexed: 07/05/2024] Open
Abstract
In this study, we investigated the role of a newly identified homozygous variant (c.1245 + 6T > C) in the CFAP61 gene in the development of multiple morphologically abnormal flagella (MMAF) in an infertile patient. Using exome sequencing, we identified this variant, which led to exon 12 skipping and the production of a truncated CFAP61 protein. Transmission electron microscopy analysis of the patient's spermatozoa revealed various flagellar abnormalities, including defective nuclear chromatin condensation, axoneme disorganization, and mitochondria embedded in residual cytoplasmic droplets. Despite a fertilization rate of 83.3% through ICSI, there was no successful pregnancy due to poor embryo quality.Our findings suggest a link between the identified CFAP61 variant and MMAF, indicating potential disruption in radial spokes' assembly or function crucial for normal ciliary motility. Furthermore, nearly half of the observed sperm heads displayed chromatin condensation defects, possibly contributing to the low blastulation rate. This case underscores the significance of genetic counseling and testing, particularly for couples dealing with infertility and MMAF. Early identification of such genetic variants can guide appropriate interventions and improve reproductive outcomes.
Collapse
Affiliation(s)
- Anne-Laure Barbotin
- UMRS1172 Development and Plasticity of the Neuroendocrine Brain, University of Lille, Lille, France
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, CHU Lille, Lille, France
| | - Angèle Boursier
- UMRS1172 Development and Plasticity of the Neuroendocrine Brain, University of Lille, Lille, France
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, CHU Lille, Lille, France
| | - Anne-Sophie Jourdain
- ULR7364 -RADEME - Maladies Rares du Développement Embryonnaire, University of Lille, Lille, France
- Institut de Génétique Médicale, CHU Lille, Avenue Oscar Lambret, F-59000, Lille, France
| | | | - Baptiste Rabat
- ULR7364 -RADEME - Maladies Rares du Développement Embryonnaire, University of Lille, Lille, France
| | - Mariam Chehimi
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, CHU Lille, Lille, France
| | - Caroline Thuillier
- Institut de Génétique Médicale, CHU Lille, Avenue Oscar Lambret, F-59000, Lille, France
| | - Jamal Ghoumid
- ULR7364 -RADEME - Maladies Rares du Développement Embryonnaire, University of Lille, Lille, France
- Clinique de Génétique Guy Fontaine, CHU Lille, Lille, France
| | - Thomas Smol
- ULR7364 -RADEME - Maladies Rares du Développement Embryonnaire, University of Lille, Lille, France.
- Institut de Génétique Médicale, CHU Lille, Avenue Oscar Lambret, F-59000, Lille, France.
| |
Collapse
|
25
|
Houston BJ, Merriner DJ, Stathatos GG, Nguyen JH, O'Connor AE, Lopes AM, Conrad DF, Baker M, Dunleavy JE, O'Bryan MK. Genetic mutation of Cep76 results in male infertility due to abnormal sperm tail composition. Life Sci Alliance 2024; 7:e202302452. [PMID: 38570187 PMCID: PMC10992998 DOI: 10.26508/lsa.202302452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
The transition zone is a specialised gate at the base of cilia/flagella, which separates the ciliary compartment from the cytoplasm and strictly regulates protein entry. We identified a potential new regulator of the male germ cell transition zone, CEP76. We demonstrated that CEP76 was involved in the selective entry and incorporation of key proteins required for sperm function and fertility into the ciliary compartment and ultimately the sperm tail. In the mutant, sperm tails were shorter and immotile as a consequence of deficits in essential sperm motility proteins including DNAH2 and AKAP4, which accumulated at the sperm neck in the mutant. Severe annulus, fibrous sheath, and outer dense fibre abnormalities were also detected in sperm lacking CEP76. Finally, we identified that CEP76 dictates annulus positioning and structure. This study suggests CEP76 as a male germ cell transition zone protein and adds further evidence to the hypothesis that the spermatid transition zone and annulus are part of the same functional structure.
Collapse
Affiliation(s)
- Brendan J Houston
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - D Jo Merriner
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - G Gemma Stathatos
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Joseph H Nguyen
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Anne E O'Connor
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Alexandra M Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology & Immunology, University of Porto, Porto, Portugal
| | - Donald F Conrad
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Mark Baker
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Jessica Em Dunleavy
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Moira K O'Bryan
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| |
Collapse
|
26
|
Esfandyari F, Raeeszadeh M, Amiri AA. Comparative Evaluation of Levamisole and Broccoli in Mitigating Testicular Oxidative Stress and Apoptotic Alterations Caused by Cadmium and Lead Exposure in Rats. Biol Trace Elem Res 2024:10.1007/s12011-024-04241-1. [PMID: 38801623 DOI: 10.1007/s12011-024-04241-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Considering the significance of heavy metals in infertility and their reduction through natural and synthetic compounds, a comparative study of broccoli and levamisole in cadmium and lead poisoning was conducted. Male Wistar rats (48 in total) were divided into 8 groups. Control, cadmium, lead, levamisole, and broccoli were administered individually to groups 1-5, while groups 6-8 received combinations. Various measurements were taken, including final weight, testicular weight, and the GSI coefficient. Sperm parameters, spermatogenesis cell count, oxidative stress biomarkers, and apoptosis indices were assessed using ELISA kits and methods in testicular tissue. The results indicated that the GSI coefficient was lowest in group 2 and highest in group 4, showing a significant difference (P < 0.001). Sperm concentration peaked in group 1 and broccoli-treated ones, while motility was highest in group 5. Testicular cell counts and Johnson score were highest in groups 1 and 2, and lowest in cadmium-exposed groups. These differences were statistically significant at P < 0.01. Enzyme activities related to oxidative stress varied. Group 2 exhibited the highest catalase (CAT) and superoxide dismutase (SOD) activities, while glutathione peroxidase (GPx) levels peaked in groups 1, 4, and 5. Malondialdehyde (MDA) concentrations were significantly reduced in the group 5 (P < 0.05). Apoptosis indices revealed that broccoli had the highest Bcl-2 levels and lowest Bax/Bcl-2 ratio, indicating its anti-apoptotic effect. Group 4 showed less efficacy compared to broccoli in protecting fertility indices. In conclusion, cadmium and lead significantly impact male fertility, while broccoli extract demonstrates promising efficacy in mitigating damage when compared to levamisole. This underscores its antioxidant and anti-apoptotic properties.
Collapse
Affiliation(s)
- Fatemeh Esfandyari
- Faculty of Veterinary Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Mahdieh Raeeszadeh
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| | - Ali Akbar Amiri
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| |
Collapse
|
27
|
Guo R, Wu H, Zhu X, Wang G, Hu K, Li K, Geng H, Xu C, Zu C, Gao Y, Tang D, Cao Y, He X. Bi-allelic variants in chromatoid body protein TDRD6 cause spermiogenesis defects and severe oligoasthenoteratozoospermia in humans. J Med Genet 2024; 61:553-565. [PMID: 38341271 DOI: 10.1136/jmg-2023-109766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND The association between the TDRD6 variants and human infertility remains unclear, as only one homozygous missense variant of TDRD6 was found to be associated with oligoasthenoteratozoospermia (OAT). METHODS Whole-exome sequencing and Sanger sequencing were employed to identify potential pathogenic variants of TDRD6 in infertile men. Histology, immunofluorescence, immunoblotting and ultrastructural analyses were conducted to clarify the structural and functional abnormalities of sperm in mutated patients. Tdrd6-knockout mice were generated using the CRISPR-Cas9 system. Total RNA-seq and single-cell RNA-seq (scRNA-seq) analyses were used to elucidate the underlying molecular mechanisms, followed by validation through quantitative RT-PCR and immunostaining. Intracytoplasmic sperm injection (ICSI) was also used to assess the efficacy of clinical treatment. RESULTS Bi-allelic TDRD6 variants were identified in five unrelated Chinese individuals with OAT, including homozygous loss-of-function variants in two consanguineous families. Notably, besides reduced concentrations and impaired motility, a significant occurrence of acrosomal hypoplasia was detected in multiple spermatozoa among five patients. Using the Tdrd6-deficient mice, we further elucidate the pivotal role of TDRD6 in spermiogenesis and acrosome identified. In addition, the mislocalisation of crucial chromatoid body components DDX4 (MVH) and UPF1 was also observed in round spermatids from patients harbouring TDRD6 variants. ScRNA-seq analysis of germ cells from a patient with TDRD6 variants revealed that TDRD6 regulates mRNA metabolism processes involved in spermatid differentiation and cytoplasmic translation. CONCLUSION Our findings strongly suggest that TDRD6 plays a conserved role in spermiogenesis and confirms the causal relationship between TDRD6 variants and human OAT. Additionally, this study highlights the unfavourable ICSI outcomes in individuals with bi-allelic TDRD6 variants, providing insights for potential clinical treatment strategies.
Collapse
Affiliation(s)
- Rui Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Huan Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Xiaoyu Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Guanxiong Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Kaiqin Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Kuokuo Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Hao Geng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Chuan Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Chenwan Zu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Yang Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Engineering Research Center of Biopreservation and Artifical Organs, Ministry of Education, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Dongdong Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Dupuis S, Girault MS, Le Beulze M, Ialy-Radio C, Bermúdez-Guzmán L, Ziyyat A, Barbaux S. The lack of Tex44 causes severe subfertility with flagellar abnormalities in male mice. Cell Mol Biol Lett 2024; 29:74. [PMID: 38750428 PMCID: PMC11094962 DOI: 10.1186/s11658-024-00587-5] [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] [Received: 01/03/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
By analyzing a mouse Interspecific Recombinant Congenic Strain (IRCS), we previously identified a quantitative trait locus (QTL), called Mafq1 on mouse chromosome 1, that is associated with male hypofertility and ultrastructural sperm abnormalities. Within this locus, we identified a new candidate gene that could be implicated in a reproductive phenotype: Tex44 (Testis-expressed protein 44). We thus performed a CRISPR/Cas9-mediated complete deletion of this gene in mice in order to study its function. Tex44-KO males were severely hypofertile in vivo and in vitro due to a drastic reduction of sperm motility which itself resulted from important morphological sperm abnormalities. Namely, Tex44-KO sperm showed a disorganized junction between the midpiece and the principal piece of the flagellum, leading to a 180° flagellar bending in this region. In addition, the loss of some axonemal microtubule doublets and outer dense fibers in the flagellum's principal piece has been observed. Our results suggest that, in mice, TEX44 is implicated in the correct set-up of the sperm flagellum during spermiogenesis and its absence leads to flagellar abnormalities and consequently to severe male hypofertility.
Collapse
Affiliation(s)
- Sophie Dupuis
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | | | - Morgane Le Beulze
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Côme Ialy-Radio
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | | | - Ahmed Ziyyat
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
- Service d'Histologie, d'Embryologie, Biologie de La Reproduction, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - Sandrine Barbaux
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France.
| |
Collapse
|
29
|
Atiakshin D, Kulchenko N, Kostin A, Ignatyuk M, Protasov A, Klabukov I, Baranovskii D, Faniev M, Korovyakova E, Chekmareva I, Buchwalow I, Tiemann M. Cyto- and Histopographic Assessment of CPA3-Positive Testicular Mast Cells in Obstructive and Non-Obstructive Azoospermia. Cells 2024; 13:833. [PMID: 38786055 PMCID: PMC11120214 DOI: 10.3390/cells13100833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Infertility is an important personal and society disease, of which the male factor represents half of all causes. One of the aspects less studied in male infertility is the immunological testicular microenvironment. Mast cells (MCs), having high potential for regulating spermatogenesis due to fine-tuning the state of the integrative buffer metabolic environment, are one of the most crucial cellular subpopulations of the testicular interstitium. One important component of the MC secretome is proteases that can act as proinflammatory agents and in extracellular matrix (ECM) remodeling. In the testis, MCs are an important cell component of the testicular interstitial tissue (TIT). However, there are still no studies addressing the analysis of a specific MC protease-carboxypeptidase A3 (CPA3)-in cases with altered spermatogenesis. The cytological and histotopographic features of testicular CPA3+ MCs were examined in a study involving 34 men with azoospermia. As revealed, in cases with non-obstructive azoospermia, a higher content of CPA3+ MCs in the TIT and migration to the microvasculature and peritubular tissue of seminiferous tubules were observed when compared with cases with obstructive azoospermia. Additionally, a high frequency of CPA3+ MCs colocalization with fibroblasts, Leydig cells, and elastic fibers was detected in cases with NOA. Thus, CPA3 seems to be of crucial pathogenetic significance in the formation of a profibrogenic background of the tissue microenvironment, which may have direct and indirect effects on spermatogenesis.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Nina Kulchenko
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Andrey Kostin
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Michael Ignatyuk
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Andrey Protasov
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia (D.B.)
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia (D.B.)
| | - Mikhail Faniev
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Elina Korovyakova
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Irina Chekmareva
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
| | - Igor Buchwalow
- RUDN University, 117198 Moscow, Russia; (N.K.); (A.K.); (M.I.); (A.P.); (M.F.); (E.K.); (I.C.); (I.B.)
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany;
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany;
| |
Collapse
|
30
|
Graziani A, Rocca MS, Vinanzi C, Masi G, Grande G, De Toni L, Ferlin A. Genetic Causes of Qualitative Sperm Defects: A Narrative Review of Clinical Evidence. Genes (Basel) 2024; 15:600. [PMID: 38790229 PMCID: PMC11120687 DOI: 10.3390/genes15050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Several genes are implicated in spermatogenesis and fertility regulation, and these genes are presently being analysed in clinical practice due to their involvement in male factor infertility (MFI). However, there are still few genetic analyses that are currently recommended for use in clinical practice. In this manuscript, we reviewed the genetic causes of qualitative sperm defects. We distinguished between alterations causing reduced sperm motility (asthenozoospermia) and alterations causing changes in the typical morphology of sperm (teratozoospermia). In detail, the genetic causes of reduced sperm motility may be found in the alteration of genes associated with sperm mitochondrial DNA, mitochondrial proteins, ion transport and channels, and flagellar proteins. On the other hand, the genetic causes of changes in typical sperm morphology are related to conditions with a strong genetic basis, such as macrozoospermia, globozoospermia, and acephalic spermatozoa syndrome. We tried to distinguish alterations approved for routine clinical application from those still unsupported by adequate clinical studies. The most important aspect of the study was related to the correct identification of subjects to be tested and the correct application of genetic tests based on clear clinical data. The correct application of available genetic tests in a scenario where reduced sperm motility and changes in sperm morphology have been observed enables the delivery of a defined diagnosis and plays an important role in clinical decision-making. Finally, clarifying the genetic causes of MFI might, in future, contribute to reducing the proportion of so-called idiopathic MFI, which might indeed be defined as a subtype of MFI whose cause has not yet been revealed.
Collapse
Affiliation(s)
- Andrea Graziani
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Maria Santa Rocca
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Cinzia Vinanzi
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Giulia Masi
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Giuseppe Grande
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| | - Luca De Toni
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
| | - Alberto Ferlin
- Department of Medicine, University of Padova, 35128 Padova, Italy; (A.G.); (G.M.); (L.D.T.)
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy; (M.S.R.); (C.V.); (G.G.)
| |
Collapse
|
31
|
Lillepea K, Juchnewitsch AG, Kasak L, Valkna A, Dutta A, Pomm K, Poolamets O, Nagirnaja L, Tamp E, Mahyari E, Vihljajev V, Tjagur S, Papadimitriou S, Riera-Escamilla A, Versbraegen N, Farnetani G, Castillo-Madeen H, Sütt M, Kübarsepp V, Tennisberg S, Korrovits P, Krausz C, Aston KI, Lenaerts T, Conrad DF, Punab M, Laan M. Toward clinical exomes in diagnostics and management of male infertility. Am J Hum Genet 2024; 111:877-895. [PMID: 38614076 PMCID: PMC11080280 DOI: 10.1016/j.ajhg.2024.03.013] [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: 12/21/2023] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 04/15/2024] Open
Abstract
Infertility, affecting ∼10% of men, is predominantly caused by primary spermatogenic failure (SPGF). We screened likely pathogenic and pathogenic (LP/P) variants in 638 candidate genes for male infertility in 521 individuals presenting idiopathic SPGF and 323 normozoospermic men in the ESTAND cohort. Molecular diagnosis was reached for 64 men with SPGF (12%), with findings in 39 genes (6%). The yield did not differ significantly between the subgroups with azoospermia (20/185, 11%), oligozoospermia (18/181, 10%), and primary cryptorchidism with SPGF (26/155, 17%). Notably, 19 of 64 LP/P variants (30%) identified in 28 subjects represented recurrent findings in this study and/or with other male infertility cohorts. NR5A1 was the most frequently affected gene, with seven LP/P variants in six SPGF-affected men and two normozoospermic men. The link to SPGF was validated for recently proposed candidate genes ACTRT1, ASZ1, GLUD2, GREB1L, LEO1, RBM5, ROS1, and TGIF2LY. Heterozygous truncating variants in BNC1, reported in female infertility, emerged as plausible causes of severe oligozoospermia. Data suggested that several infertile men may present congenital conditions with less pronounced or pleiotropic phenotypes affecting the development and function of the reproductive system. Genes regulating the hypothalamic-pituitary-gonadal axis were affected in >30% of subjects with LP/P variants. Six individuals had more than one LP/P variant, including five with two findings from the gene panel. A 4-fold increased prevalence of cancer was observed in men with genetic infertility compared to the general male population (8% vs. 2%; p = 4.4 × 10-3). Expanding genetic testing in andrology will contribute to the multidisciplinary management of SPGF.
Collapse
Affiliation(s)
- Kristiina Lillepea
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Anna-Grete Juchnewitsch
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Laura Kasak
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Anu Valkna
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Avirup Dutta
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Kristjan Pomm
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Olev Poolamets
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Erik Tamp
- Center of Pathology, Diagnostic Clinic, East Tallinn Central Hospital, 10138 Tallinn, Estonia
| | - Eisa Mahyari
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | | | - Stanislav Tjagur
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Sofia Papadimitriou
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium; Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium; Department of Biomolecular Medicine, Faculty of Medicine and Health Science, Ghent University, 9000 Ghent, Belgium
| | - Antoni Riera-Escamilla
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Andrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, 08025 Barcelona, Catalonia, Spain
| | - Nassim Versbraegen
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium; Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Ginevra Farnetani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Helen Castillo-Madeen
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Mailis Sütt
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Viljo Kübarsepp
- Department of Surgery, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia; Department of Pediatric Surgery, Clinic of Surgery, Tartu University Hospital, 51014 Tartu, Estonia
| | - Sven Tennisberg
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Paul Korrovits
- Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia
| | - Csilla Krausz
- Andrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, 08025 Barcelona, Catalonia, Spain; Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Kenneth I Aston
- Andrology and IVF Laboratory, Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Tom Lenaerts
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium; Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium; Artificial Intelligence Laboratory, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA; Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Beaverton, OR 97239, USA
| | - Margus Punab
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; Andrology Clinic, Tartu University Hospital, 50406 Tartu, Estonia; Department of Surgery, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia.
| | - Maris Laan
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia.
| |
Collapse
|
32
|
Wang Y, Chen J, Huang X, Wu B, Dai P, Zhang F, Li J, Wang L. Gene-knockout by iSTOP enables rapid reproductive disease modeling and phenotyping in germ cells of the founder generation. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1035-1050. [PMID: 38332217 DOI: 10.1007/s11427-023-2408-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/29/2023] [Indexed: 02/10/2024]
Abstract
Cytosine base editing achieves C•G-to-T•A substitutions and can convert four codons (CAA/CAG/CGA/TGG) into STOP-codons (induction of STOP-codons, iSTOP) to knock out genes with reduced mosaicism. iSTOP enables direct phenotyping in founders' somatic cells, but it remains unknown whether this works in founders' germ cells so as to rapidly reveal novel genes for fertility. Here, we initially establish that iSTOP in mouse zygotes enables functional characterization of known genes in founders' germ cells: Cfap43-iSTOP male founders manifest expected sperm features resembling human "multiple morphological abnormalities of the flagella" syndrome (i.e., MMAF-like features), while oocytes of Zp3-iSTOP female founders have no zona pellucida. We further illustrate iSTOP's utility for dissecting the functions of unknown genes with Ccdc183, observing MMAF-like features and male infertility in Ccdc183-iSTOP founders, phenotypes concordant with those of Ccdc183-KO offspring. We ultimately establish that CCDC183 is essential for sperm morphogenesis through regulating the assembly of outer dynein arms and participating in the intra-flagellar transport. Our study demonstrates iSTOP as an efficient tool for direct reproductive disease modeling and phenotyping in germ cells of the founder generation, and rapidly reveals the essentiality of Ccdc183 in fertility, thus providing a time-saving approach for validating genetic defects (like nonsense mutations) for human infertility.
Collapse
Affiliation(s)
- Yaling Wang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Jingwen Chen
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200433, China
| | - Xueying Huang
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bangguo Wu
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200433, China
| | - Peng Dai
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Feng Zhang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lingbo Wang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China.
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
33
|
Fontana L, Sirchia SM, Pesenti C, Colpi GM, Miozzo MR. Non-invasive biomarkers for sperm retrieval in non-obstructive patients: a comprehensive review. Front Endocrinol (Lausanne) 2024; 15:1349000. [PMID: 38689732 PMCID: PMC11058837 DOI: 10.3389/fendo.2024.1349000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Recent advancements in reproductive medicine have guided novel strategies for addressing male infertility, particularly in cases of non-obstructive azoospermia (NOA). Two prominent invasive interventions, namely testicular sperm extraction (TESE) and microdissection TESE (micro-TESE), have emerged as key techniques to retrieve gametes for assisted reproduction technologies (ART). Both heterogeneity and complexity of NOA pose a multifaceted challenge to clinicians, as the invasiveness of these procedures and their unpredictable success underscore the need for more precise guidance. Seminal plasma can be aptly regarded as a liquid biopsy of the male reproductive tract, encompassing secretions from the testes, epididymides, seminal vesicles, bulbourethral glands, and prostate. This fluid harbors a variety of cell-free nucleic acids, microvesicles, proteins, and metabolites intricately linked to gonadal activity. However, despite numerous investigations exploring potential biomarkers from seminal fluid, their widespread inclusion into the clinical practice remains limited. This could be partially due to the complex interplay of diverse clinical and genetic factors inherent to NOA that likely contributes to the absence of definitive biomarkers for residual spermatogenesis. It is conceivable that the integration of clinical data with biomarkers could increase the potential in predicting surgical procedure outcomes and their choice in NOA cases. This comprehensive review addresses the challenge of sperm retrieval in NOA through non-invasive biomarkers. Moreover, we delve into promising perspectives, elucidating innovative approaches grounded in multi-omics methodologies, including genomics, transcriptomics and proteomics. These cutting-edge techniques, combined with the clinical and genetics features of patients, could improve the use of biomarkers in personalized medical approaches, patient counseling, and the decision-making continuum. Finally, Artificial intelligence (AI) holds significant potential in the realm of combining biomarkers and clinical data, also in the context of identifying non-invasive biomarkers for sperm retrieval.
Collapse
Affiliation(s)
- Laura Fontana
- Medical Genetics Unit, Aziende Socio Sanitarie Territoriali (ASST) Santi Paolo e Carlo, Milan, Italy
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Silvia M. Sirchia
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pesenti
- Medical Genetics Unit, Aziende Socio Sanitarie Territoriali (ASST) Santi Paolo e Carlo, Milan, Italy
| | - Giovanni Maria Colpi
- Next Fertility Procrea, International Center for Assisted Reproductive Technology, Lugano, Switzerland
| | - Monica R. Miozzo
- Medical Genetics Unit, Aziende Socio Sanitarie Territoriali (ASST) Santi Paolo e Carlo, Milan, Italy
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
34
|
De Jonge CJ, Barratt CLR, Aitken RJ, Anderson RA, Baker P, Chan DYL, Connolly MP, Eisenberg ML, Garrido N, Jørgensen N, Kimmins S, Krausz C, McLachlan RI, Niederberger C, O’Bryan MK, Pacey A, Priskorn L, Rautakallio-Hokkanen S, Serour G, Veltman JA, Vogel DL, Vazquez-Levin MH. Current global status of male reproductive health. Hum Reprod Open 2024; 2024:hoae017. [PMID: 38699533 PMCID: PMC11065475 DOI: 10.1093/hropen/hoae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/22/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND The widespread interest in male reproductive health (MRH), fueled by emerging evidence, such as the global decline in sperm counts, has intensified concerns about the status of MRH. Consequently, there is a pressing requirement for a strategic, systematic approach to identifying critical questions, collecting pertinent information, and utilizing these data to develop evidence-based strategies. The methods for addressing these questions and the pathways toward their answers will inevitably vary based on the variations in cultural, geopolitical, and health-related contexts. To address these issues, a conjoint ESHRE and Male Reproductive Health Initiative (MRHI) Campus workshop was convened. OBJECTIVE AND RATIONALE The three objectives were: first, to assess the current state of MRH around the world; second, to identify some of the key gaps in knowledge; and, third, to examine how MRH stakeholders can collaboratively generate intelligent and effective paths forward. SEARCH METHODS Each expert reviewed and summarized the current literature that was subsequently used to provide a comprehensive overview of challenges related to MRH. OUTCOMES This narrative report is an overview of the data, opinions, and arguments presented during the workshop. A number of outcomes are presented and can be summarized by the following overarching themes: MRH is a serious global issue and there is a plethora of gaps in our understanding; there is a need for widespread international collaborative networks to undertake multidisciplinary research into fundamental issues, such as lifestyle/environmental exposure studies, and high-quality clinical trials; and there is an urgent requirement for effective strategies to educate young people and the general public to safeguard and improve MRH across diverse population demographics and resources. LIMITATIONS REASONS FOR CAUTION This was a workshop where worldwide leading experts from a wide range of disciplines presented and discussed the evidence regarding challenges related to MRH. While each expert summarized the current literature and placed it in context, the data in a number of areas are limited and/or sparse. Equally, important areas for consideration may have been missed. Moreover, there are clear gaps in our knowledge base, which makes some conclusions necessarily speculative and warranting of further study. WIDER IMPLICATIONS Poor MRH is a global issue that suffers from low awareness among the public, patients, and heathcare professionals. Addressing this will require a coordinated multidisciplinary approach. Addressing the significant number of knowledge gaps will require policy makers prioritizing MRH and its funding. STUDY FUNDING/COMPETING INTERESTS The authors would like to extend their gratitude to ESHRE for providing financial support for the Budapest Campus Workshop, as well as to Microptic S.L. (Barcelona) for kindly sponsoring the workshop. P.B. is the Director of the not-for-profit organization Global Action on Men's Health and receives fees and expenses for his work, (which includes the preparation of this manuscript). Conflicts of interest: C.J.D.J., C.L.R.B., R.A.A., P.B., M.P.C., M.L.E., N.G., N.J., C.K., AAP, M.K.O., S.R.-H., M.H.V.-L.: ESHRE Campus Workshop 2022 (Travel support-personal). C.J.D.J.: Cambridge University Press (book royalties-personal). ESHRE Annual Meeting 2022 and Yale University Panel Meeting 2023 (Travel support-personal). C.L.R.B.: Ferring and IBSA (Lecture), RBMO editor (Honorarium to support travel, etc.), ExSeed and ExScentia (University of Dundee), Bill & Melinda Gates Foundation (for research on contraception). M.P.C.: Previously received funding from pharmaceutical companies for health economic research. The funding was not in relation to this work and had no bearing on the contents of this work. No funding from other sources has been provided in relation to this work (funding was provided to his company Global Market Access Solutions). M.L.E.: Advisor to Ro, Doveras, Next, Hannah, Sandstone. C.K.: European Academy of Andrology (Past president UNPAID), S.K.: CEO of His Turn, a male fertility Diagnostic and Therapeutic company (No payments or profits to date). R.I.M.: www.healthymale.org.au (Australian Government funded not for profit in men's health sector (Employed as Medical Director 0.2 FET), Monash IVF Pty Ltd (Equity holder)). N.J.: Merck (consulting fees), Gedeon Richter (honoraria). S.R.-H.: ESHRE (Travel reimbursements). C.N.: LLC (Nursing educator); COMMIT (Core Outcomes Measures for Infertility Trials) Advisor, meeting attendee, and co-author; COMMA (Core Outcomes in Menopause) Meeting attendee, and co-author; International Federation of Gynecology and Obstetrics (FIGO) Delegate Letters and Sciences; ReproNovo, Advisory board; American Board of Urology Examiner; American Urological Association Journal subsection editor, committee member, guidelines co-author Ferring Scientific trial NexHand Chief Technology Officer, stock ownership Posterity Health Board member, stock ownership. A.P.: Economic and Social Research Council (A collaborator on research grant number ES/W001381/1). Member of an advisory committee for Merck Serono (November 2022), Member of an advisory board for Exceed Health, Speaker fees for educational events organized by Mealis Group; Chairman of the Cryos External Scientific Advisory Committee: All fees associated with this are paid to his former employer The University of Sheffield. Trustee of the Progress Educational Trust (Unpaid). M.K.O.: National Health and Medical Research Council and Australian Research Council (Funding for research of the topic of male fertility), Bill and Melinda Gates Foundation (Funding aimed at the development of male gamete-based contraception), Medical Research Future Fund (Funding aimed at defining the long-term consequences of male infertility). M.H.V.-L.: Department of Sexual and Reproductive Health and Research (SRH)/Human Reproduction Programme (HRP) Research Project Panel RP2/WHO Review Member; MRHI (Core Group Member), COMMIT (member), EGOI (Member); Human Reproduction (Associate Editor), Fertility and Sterility (Editor), AndroLATAM (Founder and Coordinator).
Collapse
Affiliation(s)
- Christopher J De Jonge
- Department of Urology, University of Minnesota Medical Center, University of Minnesota, Minneapolis, MN, USA
| | - Christopher L R Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - R John Aitken
- Discipline of Biological Sciences, School of Environment and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Newcastle, Australia
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - David Y L Chan
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
| | - Mark P Connolly
- Health Economics, Global Market Access Solutions LLC, Mooresville, NC, USA
- University Medical Center Groningen, Groningen, The Netherlands
| | - Michael L Eisenberg
- Department of Urology and Obstetrics & Gynecology, Stanford University, Stanford, CA, USA
| | - Nicolas Garrido
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences, ‘Mario Serio’, University of Florence, University Hospital of Careggi (AOUC), Florence, Italy
| | - Robert I McLachlan
- Hudson Institute of Medical Research, Monash University, Melbourne, Australia
- Monash IVF Group, Cremorne, Australia
| | - Craig Niederberger
- Clarence C. Department of Urology, University of Illinois Chicago (UIC), College of Medicine, Department of Bioengineering, UIC College of Engineering, Chicago, IL,USA
| | - Moira K O’Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Allan Pacey
- Faculty of Biology, Medicine and Health, Core Technology Facility, University of Manchester, Manchester, UK
| | - Lærke Priskorn
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Gamal Serour
- The International Islamic Center for Population Studies and Research, Al-Azhar University, Maadi, Cairo, Egypt
- Egyptian IVF Center, Maadi, Cairo, Egypt
| | - Joris A Veltman
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Donna L Vogel
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Mónica H Vazquez-Levin
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina—Fundación IBYME, Buenos Aires, Argentina
| |
Collapse
|
35
|
Olszewska M, Malcher A, Stokowy T, Pollock N, Berman AJ, Budkiewicz S, Kamieniczna M, Jackowiak H, Suszynska-Zajczyk J, Jedrzejczak P, Yatsenko AN, Kurpisz M. Effects of Tcte1 knockout on energy chain transportation and spermatogenesis: implications for male infertility. Hum Reprod Open 2024; 2024:hoae020. [PMID: 38650655 PMCID: PMC11035007 DOI: 10.1093/hropen/hoae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/08/2024] [Indexed: 04/25/2024] Open
Abstract
STUDY QUESTION Is the Tcte1 mutation causative for male infertility? SUMMARY ANSWER Our collected data underline the complex and devastating effect of the single-gene mutation on the testicular molecular network, leading to male reproductive failure. WHAT IS KNOWN ALREADY Recent data have revealed mutations in genes related to axonemal dynein arms as causative for morphology and motility abnormalities in spermatozoa of infertile males, including dysplasia of fibrous sheath (DFS) and multiple morphological abnormalities in the sperm flagella (MMAF). The nexin-dynein regulatory complex (N-DRC) coordinates the dynein arm activity and is built from the DRC1-DRC7 proteins. DRC5 (TCTE1), one of the N-DRC elements, has already been reported as a candidate for abnormal sperm flagella beating; however, only in a restricted manner with no clear explanation of respective observations. STUDY DESIGN SIZE DURATION Using the CRISPR/Cas9 genome editing technique, a mouse Tcte1 gene knockout line was created on the basis of the C57Bl/6J strain. The mouse reproductive potential, semen characteristics, testicular gene expression levels, sperm ATP, and testis apoptosis level measurements were then assessed, followed by visualization of N-DRC proteins in sperm, and protein modeling in silico. Also, a pilot genomic sequencing study of samples from human infertile males (n = 248) was applied for screening of TCTE1 variants. PARTICIPANTS/MATERIALS SETTING METHODS To check the reproductive potential of KO mice, adult animals were crossed for delivery of three litters per caged pair, but for no longer than for 6 months, in various combinations of zygosity. All experiments were performed for wild-type (WT, control group), heterozygous Tcte1+/- and homozygous Tcte1-/- male mice. Gross anatomy was performed on testis and epididymis samples, followed by semen analysis. Sequencing of RNA (RNAseq; Illumina) was done for mice testis tissues. STRING interactions were checked for protein-protein interactions, based on changed expression levels of corresponding genes identified in the mouse testis RNAseq experiments. Immunofluorescence in situ staining was performed to detect the N-DRC complex proteins: Tcte1 (Drc5), Drc7, Fbxl13 (Drc6), and Eps8l1 (Drc3) in mouse spermatozoa. To determine the amount of ATP in spermatozoa, the luminescence level was measured. In addition, immunofluorescence in situ staining was performed to check the level of apoptosis via caspase 3 visualization on mouse testis samples. DNA from whole blood samples of infertile males (n = 137 with non-obstructive azoospermia or cryptozoospermia, n = 111 samples with a spectrum of oligoasthenoteratozoospermia, including n = 47 with asthenozoospermia) was extracted to perform genomic sequencing (WGS, WES, or Sanger). Protein prediction modeling of human-identified variants and the exon 3 structure deleted in the mouse knockout was also performed. MAIN RESULTS AND THE ROLE OF CHANCE No progeny at all was found for the homozygous males which were revealed to have oligoasthenoteratozoospermia, while heterozygous animals were fertile but manifested oligozoospermia, suggesting haploinsufficiency. RNA-sequencing of the testicular tissue showed the influence of Tcte1 mutations on the expression pattern of 21 genes responsible for mitochondrial ATP processing or linked with apoptosis or spermatogenesis. In Tcte1-/- males, the protein was revealed in only residual amounts in the sperm head nucleus and was not transported to the sperm flagella, as were other N-DRC components. Decreased ATP levels (2.4-fold lower) were found in the spermatozoa of homozygous mice, together with disturbed tail:midpiece ratios, leading to abnormal sperm tail beating. Casp3-positive signals (indicating apoptosis) were observed in spermatogonia only, at a similar level in all three mouse genotypes. Mutation screening of human infertile males revealed one novel and five ultra-rare heterogeneous variants (predicted as disease-causing) in 6.05% of the patients studied. Protein prediction modeling of identified variants revealed changes in the protein surface charge potential, leading to disruption in helix flexibility or its dynamics, thus suggesting disrupted interactions of TCTE1 with its binding partners located within the axoneme. LARGE SCALE DATA All data generated or analyzed during this study are included in this published article and its supplementary information files. RNAseq data are available in the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE207805. The results described in the publication are based on whole-genome or exome sequencing data which includes sensitive information in the form of patient-specific germline variants. Information regarding such variants must not be shared publicly following European Union legislation, therefore access to raw data that support the findings of this study are available from the corresponding author upon reasonable request. LIMITATIONS REASONS FOR CAUTION In the study, the in vitro fertilization performance of sperm from homozygous male mice was not checked. WIDER IMPLICATIONS OF THE FINDINGS This study contains novel and comprehensive data concerning the role of TCTE1 in male infertility. The TCTE1 gene is the next one that should be added to the 'male infertility list' because of its crucial role in spermatogenesis and proper sperm functioning. STUDY FUNDING/COMPETING INTERESTS This work was supported by National Science Centre in Poland, grants no.: 2015/17/B/NZ2/01157 and 2020/37/B/NZ5/00549 (to M.K.), 2017/26/D/NZ5/00789 (to A.M.), and HD096723, GM127569-03, NIH SAP #4100085736 PA DoH (to A.N.Y.). The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Collapse
Affiliation(s)
- Marta Olszewska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Agnieszka Malcher
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Stokowy
- Scientific Computing Group, IT Division, University of Bergen, Bergen, Norway
| | - Nijole Pollock
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrea J Berman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sylwia Budkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Hanna Jackowiak
- Department of Histology and Embryology, Poznan University of Life Sciences, Poznan, Poland
| | | | - Piotr Jedrzejczak
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alexander N Yatsenko
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maciej Kurpisz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
36
|
Al Dala Ali M, Longepied G, Nicolet A, Metzler-Guillemain C, Mitchell MJ. Spermatozoa in mice lacking the nucleoporin NUP210L show defects in head shape and motility but not in nuclear compaction or histone replacement. Clin Genet 2024; 105:364-375. [PMID: 38129135 DOI: 10.1111/cge.14468] [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: 10/26/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Biallelic loss-of-function mutation of NUP210L, encoding a testis-specific nucleoporin, has been reported in an infertile man whose spermatozoa show uncondensed heads and histone retention. Mice with a homozygous transgene intronic insertion in Nup210l were infertile but spermatozoa had condensed heads. Expression from this insertion allele is undefined, however, and residual NUP210L production could underlie the milder phenotype. To resolve this issue, we have created Nup210lem1Mjmm , a null allele of Nup210l, in the mouse. Nup210lem1Mjmm homozygotes show uniform mild anomalies of sperm head morphology and decreased motility, but nuclear compaction and histone removal appear unaffected. Thus, our mouse model does not support that NUP210L loss alone blocks spermatid nuclear compaction. Re-analyzing the patient's exome data, we identified a rare, potentially pathogenic, heterozygous variant in nucleoporin gene NUP153 (p.Pro485Leu), and showed that, in mouse and human, NUP210L and NUP153 colocalize at the caudal nuclear pole in elongating spermatids and spermatozoa. Unexpectedly, in round spermatids, NUP210L and NUP153 localisation differs between mouse (nucleoplasm) and human (nuclear periphery). Our data suggest two explanations for the increased phenotypic severity associated with NUP210L loss in human compared to mouse: a genetic variant in human NUP153 (p.Pro485Leu), and inter-species divergence in nuclear pore function in round spermatids.
Collapse
Affiliation(s)
- Maha Al Dala Ali
- Aix Marseille University, Inserm, MMG, Marseille, France
- College of Medicine, Al-Iraqia University, Baghdad, Iraq
| | - Guy Longepied
- Aix Marseille University, Inserm, MMG, Marseille, France
| | - Aurore Nicolet
- Aix Marseille University, Inserm, MMG, Marseille, France
| | - Catherine Metzler-Guillemain
- Aix Marseille University, Inserm, MMG, Marseille, France
- AP-HM, Pôle Femmes-Parents-Enfants, Centre Clinico-biologique AMP-CECOS, Marseille Cedex 5, France
| | | |
Collapse
|
37
|
Montjean D, Beaumont M, Natiq A, Louanjli N, Hazout A, Miron P, Liehr T, Cabry R, Ratbi I, Benkhalifa M. Genome and Epigenome Disorders and Male Infertility: Feedback from 15 Years of Clinical and Research Experience. Genes (Basel) 2024; 15:377. [PMID: 38540436 PMCID: PMC10970370 DOI: 10.3390/genes15030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 06/14/2024] Open
Abstract
Infertility affects around 20% of couples of reproductive age; however, in some societies, as many as one-third of couples are unable to conceive. Different factors contribute to the decline of male fertility, such us environmental and professional exposure to endocrine disruptors, oxidative stress, and life habits with the risk of de novo epigenetics dysregulation. Since the fantastic development of new "omes and omics" technologies, the contribution of inherited or de novo genomes and epigenome disorders to male infertility have been further elucidated. Many other techniques have become available to andrology laboratories for the investigation of genome and epigenome integrity and the maturation and the competency of spermatozoa. All these new methods of assessment are highlighting the importance of genetics and epigenetics investigation for assisted reproduction pathology and for supporting professionals in counselling patients and proposing different management strategies for male infertility. This aims to improve clinical outcomes while minimizing the risk of genetics or health problems at birth.
Collapse
Affiliation(s)
- Debbie Montjean
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Marion Beaumont
- Genetics Department, Eylau/Unilabs Laboratory, 92110 Clichy, France;
| | - Abdelhafid Natiq
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- National Laboratory Mohammed VI, Mohammed VI Foundation of Casablanca, Casablanca, Morocco
| | | | - Andre Hazout
- Andro-Genetics Unit, Labomac, Casablanca, Morocco (A.H.)
| | - Pierre Miron
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Thomas Liehr
- Institute für Humangenetik, Universitätsklinikum Jena, Friedrich Schiller Universität, 07743 Jena, Germany
| | - Rosalie Cabry
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| | - Ilham Ratbi
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- Medical Genetics Unit, Ibn Sina University Hospital Center, Rabat, Morocco
| | - Moncef Benkhalifa
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| |
Collapse
|
38
|
Zhu X, Liu L, Tian S, Zhao G, Zhi E, Chen Q, Zhang F, Zhang A, Tang S, Liu C. Deleterious variant in FAM71D cause male infertility with asthenoteratospermia. Mol Genet Genomics 2024; 299:35. [PMID: 38489045 DOI: 10.1007/s00438-024-02117-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/24/2024] [Indexed: 03/17/2024]
Abstract
Asthenoteratospermia is a significant cause of male infertility. FAM71D (Family with sequence similarity 71, member D), as a novel protein exclusively expressed in the testis, has been found to be associated with sperm motility. However, the association of FAM71D mutation with male infertility has yet to be examined. Here, we conducted whole-exome sequencing and identified a homozygous missense mutation c.440G > A (p. Arg147Gln) of FAM71D in an asthenoteratospermia-affected man from a consanguineous family. The FAM71D variant is extremely rare in human population genome databases and predicted to be deleterious by multiple bioinformatics tools. Semen analysis indicated decreased sperm motility and obvious morphological abnormalities in sperm cells from the FAM71D-deficient man. Immunofluorescence assays revealed that the identified FAM71D mutation had an important influence on the assembly of sperm structure-related proteins. Furthermore, intra-cytoplasmic sperm injection (ICSI) treatment performed on the infertile man with FAM71D variant achieved a satisfactory outcome. Overall, our study identified FAM71D as a novel causative gene for male infertility with asthenoteratospermia, for which ICSI treatment may be suggested to acquire good prognosis. All these findings will provide effective guidance for genetic counselling and assisted reproduction treatments of asthenoteratospermia-affected subjects.
Collapse
Affiliation(s)
- Xiaobin Zhu
- Department of Gynecology and Obstetrics, Reproductive Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liu Liu
- Obstetrics and Gynecology Hospital, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
- Department of Computational Biology, School of Life Science, Fudan University, Shanghai, 200438, China
| | - Shixiong Tian
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200438, China
| | - Guijun Zhao
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200040, China
| | - Erlei Zhi
- Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200940, China
| | - Qian Chen
- Department of Gynecology and Obstetrics, Reproductive Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Feng Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Aijun Zhang
- Department of Gynecology and Obstetrics, Reproductive Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shuyan Tang
- Obstetrics and Gynecology Hospital, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China.
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200438, China.
| | - Chunyu Liu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
39
|
Muroňová J, Kherraf ZE, Giordani E, Lambert E, Eckert S, Cazin C, Amiri-Yekta A, Court M, Chevalier G, Martinez G, Neirijnck Y, Kühne F, Wehrli L, Klena N, Hamel V, De Macedo L, Escoffier J, Guichard P, Coutton C, Mustapha SFB, Kharouf M, Bouin AP, Zouari R, Thierry-Mieg N, Nef S, Geimer S, Loeuillet C, Ray PF, Arnoult C. Lack of CCDC146, a ubiquitous centriole and microtubule-associated protein, leads to non-syndromic male infertility in human and mouse. eLife 2024; 12:RP86845. [PMID: 38441556 PMCID: PMC10942651 DOI: 10.7554/elife.86845] [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: 03/07/2024] Open
Abstract
From a cohort of 167 infertile patients suffering from multiple morphological abnormalities of the flagellum (MMAF), pathogenic bi-allelic mutations were identified in the CCDC146 gene. In somatic cells, CCDC146 is located at the centrosome and at multiple microtubule-related organelles during mitotic division, suggesting that it is a microtubule-associated protein (MAP). To decipher the molecular pathogenesis of infertility associated with CCDC146 mutations, a Ccdc146 knock-out (KO) mouse line was created. KO male mice were infertile, and sperm exhibited a phenotype identical to CCDC146 mutated patients. CCDC146 expression starts during late spermiogenesis. In the spermatozoon, the protein is conserved but is not localized to centrioles, unlike in somatic cells, rather it is present in the axoneme at the level of microtubule doublets. Expansion microscopy associated with the use of the detergent sarkosyl to solubilize microtubule doublets suggests that the protein may be a microtubule inner protein (MIP). At the subcellular level, the absence of CCDC146 impacted all microtubule-based organelles such as the manchette, the head-tail coupling apparatus (HTCA), and the axoneme. Through this study, a new genetic cause of infertility and a new factor in the formation and/or structure of the sperm axoneme were characterized.
Collapse
Affiliation(s)
- Jana Muroňová
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Zine Eddine Kherraf
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- UM GI-DPI, CHU Grenoble AlpesGrenobleFrance
| | - Elsa Giordani
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Emeline Lambert
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Simon Eckert
- Cell Biology/ Electron Microscopy, University of BayreuthBayreuthGermany
| | - Caroline Cazin
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- UM GI-DPI, CHU Grenoble AlpesGrenobleFrance
| | - Amir Amiri-Yekta
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECRTehranIslamic Republic of Iran
| | - Magali Court
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Geneviève Chevalier
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Guillaume Martinez
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- UM de Génétique Chromosomique, Hôpital Couple-Enfant, CHU Grenoble AlpesGrenobleFrance
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva Medical SchoolGenevaSwitzerland
| | - Francoise Kühne
- Department of Genetic Medicine and Development, University of Geneva Medical SchoolGenevaSwitzerland
| | - Lydia Wehrli
- Department of Genetic Medicine and Development, University of Geneva Medical SchoolGenevaSwitzerland
| | - Nikolai Klena
- University of Geneva, Department of Molecular and Cellular Biology, Sciences IIIGenevaSwitzerland
| | - Virginie Hamel
- University of Geneva, Department of Molecular and Cellular Biology, Sciences IIIGenevaSwitzerland
| | - Lisa De Macedo
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Jessica Escoffier
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Paul Guichard
- University of Geneva, Department of Molecular and Cellular Biology, Sciences IIIGenevaSwitzerland
| | - Charles Coutton
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- UM de Génétique Chromosomique, Hôpital Couple-Enfant, CHU Grenoble AlpesGrenobleFrance
| | | | - Mahmoud Kharouf
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain NordTunisTunisia
| | - Anne-Pacale Bouin
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Raoudha Zouari
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain NordTunisTunisia
| | - Nicolas Thierry-Mieg
- Laboratoire TIMC/MAGe, CNRS UMR 5525, Pavillon Taillefer, Faculté de MedecineLa TroncheFrance
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical SchoolGenevaSwitzerland
| | - Stefan Geimer
- Cell Biology/ Electron Microscopy, University of BayreuthBayreuthGermany
| | - Corinne Loeuillet
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| | - Pierre F Ray
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
- UM GI-DPI, CHU Grenoble AlpesGrenobleFrance
| | - Christophe Arnoult
- Institute for Advanced Biosciences (IAB), INSERM 1209GrenobleFrance
- Institute for Advanced Biosciences (IAB), CNRS UMR 5309GrenobleFrance
- Institute for Advanced Biosciences (IAB), Université Grenoble AlpesGrenobleFrance
| |
Collapse
|
40
|
Sieper MH, Gaikwad AS, Fros M, Weber P, Di Persio S, Oud MS, Kliesch S, Neuhaus N, Stallmeyer B, Tüttelmann F, Wyrwoll MJ. Scrutinizing the human TEX genes in the context of human male infertility. Andrology 2024; 12:570-584. [PMID: 37594251 DOI: 10.1111/andr.13511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Infertility affects around 15% of all couples worldwide and is increasingly linked to variants in genes specifically expressed in the testis. Well-established causes of male infertility include pathogenic variants in the genes TEX11, TEX14, and TEX15, while few studies have recently reported variants in TEX13B, TEX13C, FAM9A (TEX39A), and FAM9B (TEX39B). OBJECTIVES We aimed at screening for novel potential candidate genes among the human TEX ("testis expressed") genes as well as verifying previously described disease associations in this set of genes. MATERIALS AND METHODS To this end, we screened the exome sequencing data of 1305 men, including 1056 crypto- and azoospermic individuals, and determined cell-specific expression by analyzing testis-specific single-cell RNA sequencing data for genes with identified variants. To investigate the overarching role in male fertility, we generated testis-specific knockdown (KD) models of all 10 orthologous TEX genes in Drosophila melanogaster. RESULTS We detected rare potential disease-causing variants in TEX10, TEX13A, TEX13B, TEX13C, TEX13D, ZFAND3 (TEX27), TEX33, FAM9A (TEX39A), and FAM9B (TEX39B), in 28 infertile men, of which 15 men carried variants in TEX10, TEX27, and TEX33. The KD of TEX2, TEX9, TEX10, TEX13, ZFAND3 (TEX27), TEX28, TEX30, NFX1 (TEX42), TEX261, and UTP4 (TEX292) in Drosophila resulted in normal fertility. DISCUSSION Based on our findings, the autosomal dominant predicted genes TEX10 and ZFAND3 (TEX27) and the autosomal recessive predicted gene TEX33, which all three are conceivably required for germ cell maturation, were identified as novel potential candidate genes for human non-obstructive azoospermia. We additionally identified hemizygous loss-of-function (LoF) variants in TEX13B, TEX13C, and FAM9A (TEX39A) as unlikely monogenic culprits of male infertility as LoF variants were also found in control men. CONCLUSION Our findings concerning the X-linked genes TEX13B, TEX13C, and FAM9A (TEX39A) contradict previous reports and will decrease false-positive reports in genetic diagnostics of azoospermic men.
Collapse
Affiliation(s)
- Marie H Sieper
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Avinash S Gaikwad
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Marion Fros
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Philipp Weber
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Manon S Oud
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Birgit Stallmeyer
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| |
Collapse
|
41
|
Song Y, Guo J, Zhou Y, Wei X, Li J, Zhang G, Wang H. A loss-of-function variant in ZCWPW1 causes human male infertility with sperm head defect and high DNA fragmentation. Reprod Health 2024; 21:18. [PMID: 38310235 PMCID: PMC10837985 DOI: 10.1186/s12978-024-01746-9] [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: 09/18/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Male infertility is a global health issue. The more causative genes related to human male infertility should be further explored. The essential role of Zcwpw1 in male mouse fertility has been established and the role of ZCWPW1 in human reproduction needs further investigation to verify. METHODS An infertile man with oligoasthenoteratozoospermia phenotype and his parents were recruited from West China Second University Hospital, Sichuan University. A total of 200 healthy Han Chinese volunteers without any evidence of infertility were recruited as normal controls, while an additional 150 infertile individuals were included to assess the prevalence of ZCWPW1 variants in a sporadic male sterile population. The causative gene variant was identified by Whole-exome sequencing and Sanger sequencing. The phenotype of the oligoasthenoteratozoospermia was determined by Papanicolaou staining, immunofluorescence staining and electron microscope. In-vitro experiments, western blot and in-silicon analysis were applied to assess the pathogenicity of the identified variant. Additionally, we examined the influence of the variant on the DNA fragmentation and DNA repair capability by Sperm Chromatin Dispersion and Neutral Comet Assay. RESULTS The proband exhibits a phenotype of oligoasthenoteratozoospermia, his spermatozoa show head defects by semen examination, Papanicolaou staining and electron microscope assays. Whole-exome sequencing and Sanger sequencing found the proband carries a homozygous ZCWPW1 variant (c.1064C > T, p. P355L). Immunofluorescence analysis shows a significant decrease in ZCWPW1 expression in the proband's sperm. By exogenous expression with ZCWPW1 mutant plasmid in vitro, the obvious declined expression of ZCWPW1 with the mutation is validated in HEK293T. After being treated by hydroxyurea, MUT-ZCWPW1 transfected cells and empty vector transfected cells have a higher level of γ-H2AX, increased tail DNA and reduced H3K9ac level than WT-ZCWPW1 transfected cells. Furthermore, the Sperm Chromatin Dispersion assay revealed the proband's spermatozoa have high DNA fragmentation. CONCLUSIONS It is the first report that a novel homozygous missense mutation in ZCWPW1 caused human male infertility with sperm head defects and high DNA fragmentation. This finding enriches the gene variant spectrum and etiology of oligoasthenoteratozoospermia.
Collapse
Affiliation(s)
- Yuelin Song
- Department of Obstetrics and Gynaecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Juncen Guo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yanling Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xingjian Wei
- Department of Obstetrics and Gynaecology, Southwest Medical University, Luzhou, 646000, China
| | - Jianlan Li
- Child Healthcare Department, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610000, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Guohui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610000, China.
| | - Hongjing Wang
- Department of Obstetrics and Gynaecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
42
|
Kimmins S, Anderson RA, Barratt CLR, Behre HM, Catford SR, De Jonge CJ, Delbes G, Eisenberg ML, Garrido N, Houston BJ, Jørgensen N, Krausz C, Lismer A, McLachlan RI, Minhas S, Moss T, Pacey A, Priskorn L, Schlatt S, Trasler J, Trasande L, Tüttelmann F, Vazquez-Levin MH, Veltman JA, Zhang F, O'Bryan MK. Frequency, morbidity and equity - the case for increased research on male fertility. Nat Rev Urol 2024; 21:102-124. [PMID: 37828407 DOI: 10.1038/s41585-023-00820-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/14/2023]
Abstract
Currently, most men with infertility cannot be given an aetiology, which reflects a lack of knowledge around gamete production and how it is affected by genetics and the environment. A failure to recognize the burden of male infertility and its potential as a biomarker for systemic illness exists. The absence of such knowledge results in patients generally being treated as a uniform group, for whom the strategy is to bypass the causality using medically assisted reproduction (MAR) techniques. In doing so, opportunities to prevent co-morbidity are missed and the burden of MAR is shifted to the woman. To advance understanding of men's reproductive health, longitudinal and multi-national centres for data and sample collection are essential. Such programmes must enable an integrated view of the consequences of genetics, epigenetics and environmental factors on fertility and offspring health. Definition and possible amelioration of the consequences of MAR for conceived children are needed. Inherent in this statement is the necessity to promote fertility restoration and/or use the least invasive MAR strategy available. To achieve this aim, protocols must be rigorously tested and the move towards personalized medicine encouraged. Equally, education of the public, governments and clinicians on the frequency and consequences of infertility is needed. Health options, including male contraceptives, must be expanded, and the opportunities encompassed in such investment understood. The pressing questions related to male reproductive health, spanning the spectrum of andrology are identified in the Expert Recommendation.
Collapse
Affiliation(s)
- Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- The Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- The Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, Quebec, Canada
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Christopher L R Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Hospital, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sarah R Catford
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | | | - Geraldine Delbes
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Sante Biotechnologie, Laval, Quebec, Canada
| | - Michael L Eisenberg
- Department of Urology and Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Nicolas Garrido
- IVI Foundation, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Brendan J Houston
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia
| | - Niels Jørgensen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences, 'Mario Serio', University of Florence, University Hospital of Careggi Florence, Florence, Italy
| | - Ariane Lismer
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Robert I McLachlan
- Hudson Institute of Medical Research and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
- Monash IVF Group, Richmond, Victoria, Australia
| | - Suks Minhas
- Department of Surgery and Cancer Imperial, London, UK
| | - Tim Moss
- Healthy Male and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Allan Pacey
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Lærke Priskorn
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stefan Schlatt
- Centre for Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jacquetta Trasler
- Departments of Paediatrics, Human Genetics and Pharmacology & Therapeutics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Leonardo Trasande
- Center for the Investigation of Environmental Hazards, Department of Paediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Mónica Hebe Vazquez-Levin
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Fundación IBYME, Buenos Aires, Argentina
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
43
|
Li Y, Li Q, Wu L, Wang H, Shi H, Yang C, Gu Y, Li J, Ji Z. SperMD: the expression atlas of sperm maturation. BMC Bioinformatics 2024; 25:29. [PMID: 38233783 PMCID: PMC10792849 DOI: 10.1186/s12859-024-05631-x] [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: 05/02/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
The impairment of sperm maturation is one of the major pathogenic factors in male subfertility, a serious medical and social problem affecting millions of global couples. Regrettably, the existing research on sperm maturation is slow, limited, and fragmented, largely attributable to the lack of a global molecular view. To fill the data gap, we newly established a database, namely the Sperm Maturation Database (SperMD, http://bio-add.org/SperMD ). SperMD integrates heterogeneous multi-omics data (170 transcriptomes, 91 proteomes, and five human metabolomes) to illustrate the transcriptional, translational, and metabolic manifestations during the entire lifespan of sperm maturation. These data involve almost all crucial scenarios related to sperm maturation, including the tissue components of the epididymal microenvironment, cell constituents of tissues, different pathological states, and so on. To the best of our knowledge, SperMD could be one of the limited repositories that provide focused and comprehensive information on sperm maturation. Easy-to-use web services are also implemented to enhance the experience of data retrieval and molecular comparison between humans and mice. Furthermore, the manuscript illustrates an example application demonstrated to systematically characterize novel gene functions in sperm maturation. Nevertheless, SperMD undertakes the endeavor to integrate the islanding omics data, offering a panoramic molecular view of how the spermatozoa gain full reproductive abilities. It will serve as a valuable resource for the systematic exploration of sperm maturation and for prioritizing the biomarkers and targets for precise diagnosis and therapy of male subfertility.
Collapse
Affiliation(s)
- Yifan Li
- School of Informatics, National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Qianying Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Lvying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Haiyan Wang
- Shandong Epihealth Biotech Ltd., Yantai, China
| | - Hui Shi
- College of Life Science, Yantai University, Yantai, China
| | - Chenhui Yang
- School of Informatics, National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Yiqun Gu
- Institute of science and technology, National Health Commission, Beijing, China.
| | - Jianyuan Li
- Shandong Epihealth Biotech Ltd., Yantai, China.
- Institute of science and technology, National Health Commission, Beijing, China.
| | - Zhiliang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
44
|
Daniel-Carlier N, Castille J, Passet B, Vilotte M, Le Danvic C, Jaffrezic F, Beauvallet C, Péchoux C, Capitan A, Vilotte JL. Targeted mutation and inactivation of the kinesin light chain 3 protein-encoding gene have no impact on mouse fertility†. Biol Reprod 2024; 110:78-89. [PMID: 37776549 DOI: 10.1093/biolre/ioad131] [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: 10/02/2023] Open
Abstract
The kinesin light chain 3 protein (KLC3) is the only member of the kinesin light chain protein family that was identified in post-meiotic mouse male germ cells. It plays a role in the formation of the sperm midpiece through its association with both spermatid mitochondria and outer dense fibers (ODF). Previous studies showed a significant correlation between its expression level and sperm motility and quantitative semen parameters in humans, while the overexpression of a KLC3-mutant protein unable to bind ODF also affected the same traits in mice. To further assess the role of KLC3 in fertility, we used CRISPR/Cas9 genome editing in mice and investigated the phenotypes induced by the invalidation of the gene or of a functional domain of the protein. Both approaches gave similar results, i.e. no detectable change in male or female fertility. Testis histology, litter size and sperm count were not altered. Apart from the line-dependent alterations of Klc3 mRNA levels, testicular transcriptome analysis did not reveal any other changes in the genes tested. Western analysis supported the absence of KLC3 in the gonads of males homozygous for the inactivating mutation and a strong decrease in expression in males homozygous for the allele lacking one out of the five tetratricopeptide repeats. Overall, these observations raise questions about the supposedly critical role of this kinesin in reproduction, at least in mice where its gene mutation or inactivation did not translate into fertility impairment.
Collapse
Affiliation(s)
- Nathalie Daniel-Carlier
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Johan Castille
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Bruno Passet
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Marthe Vilotte
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Christelle Le Danvic
- UVSQ, INRAE, BREED, Université Paris-Saclay, Eliance, 78350 Jouy-en-Josas, France
| | - Florence Jaffrezic
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Christian Beauvallet
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Christine Péchoux
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Aurélien Capitan
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, 78350, Jouy-en-Josas, France
| |
Collapse
|
45
|
Zimmer J, Mueller L, Frank-Herrmann P, Rehnitz J, Dietrich JE, Bettendorf M, Strowitzki T, Krivega M. Low androgen signaling rescues genome integrity with innate immune response by reducing fertility in humans. Cell Death Dis 2024; 15:30. [PMID: 38212646 PMCID: PMC10784536 DOI: 10.1038/s41419-023-06397-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024]
Abstract
Development of the gonads under complex androgen regulation is critical for germ cells specification. In this work we addressed the relationship between androgens and genomic integrity determining human fertility. We used different study groups: individuals with Differences of Sex Development (DSD), including Complete Androgen Insensitivity Syndrome (CAIS) due to mutated androgen receptor (AR), and men with idiopathic nonobstructive azoospermia. Both showed genome integrity status influenced by androgen signaling via innate immune response activation in blood and gonads. Whole proteome analysis connected low AR to interleukin-specific gene expression, while compromised genome stability and tumorigenesis were also supported by interferons. AR expression was associated with predominant DNA damage phenotype, that eliminated AR-positive Sertoli cells as the degeneration of gonads increased. Low AR contributed to resistance from the inhibition of DNA repair in primary leukocytes. Downregulation of androgen promoted apoptosis and specific innate immune response with higher susceptibility in cells carrying genomic instability.
Collapse
Affiliation(s)
- J Zimmer
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - L Mueller
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - P Frank-Herrmann
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - J Rehnitz
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - J E Dietrich
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - M Bettendorf
- Division of Pediatric Endocrinology, Children's Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - T Strowitzki
- Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany
| | - M Krivega
- Research Group of Gonadal Differentiation and Embryonic Development, Department of Gynecological Endocrinology & Fertility Disorders, Women Hospital, University of Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
46
|
Fallara G, Pozzi E, Belladelli F, Boeri L, Capogrosso P, Corona G, D'Arma A, Alfano M, Montorsi F, Salonia A. A Systematic Review and Meta-analysis on the Impact of Infertility on Men's General Health. Eur Urol Focus 2024; 10:98-106. [PMID: 37573151 DOI: 10.1016/j.euf.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/10/2023] [Accepted: 07/29/2023] [Indexed: 08/14/2023]
Abstract
CONTEXT Male infertility has been associated with increased morbidity and mortality. OBJECTIVE To perform a systematic review and meta-analysis to provide the most critical evidence on the association between infertility and the risk of incident comorbidities in males. EVIDENCE ACQUISITION A systematic review and meta-analysis was performed according to the Meta-analysis of Observational Studies in Epidemiology and Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines, and registered on PROSPERO. All published studies on infertile versus fertile men regarding overall mortality and risks of cancer, diabetes, and cardiovascular events were selected from a database search on PubMed, EMBASE, Google Scholar, and Cochrane. Forest plot and quasi-individual patient data meta-analysis were used for pooled analyses. A risk of bias was assessed using the ROBINS-E tool. EVIDENCE SYNTHESIS Overall, an increased risk of death from any cause was found for infertile men (hazard risk [HR] 1.37, [95% confidence interval {CI} 1.04-1.81], p = 0.027), and a 30-yr survival probability of 91.0% (95% CI 89.6-92.4%) was found for infertile versus 95.9% (95% CI 95.3-96.4%) for fertile men (p < 0.001). An increased risk emerged of being diagnosed with testis cancer (relative risk [RR] 1.86 [95% CI 1.41-2.45], p < 0.001), melanoma (RR 1.30 [95% CI 1.08-1.56], p = 0.006), and prostate cancer (RR 1.66 [95% CI 1.06-2.61], p < 0.001). As well, an increased risk of diabetes (HR 1.39 [95% CI 1.09-1.71], p = 0.008), with a 30-yr probability of diabetes of 25.0% (95% CI 21.1-26.9%) for infertile versus 17.1% (95% CI 16.1-18.1%) for fertile men (p < 0.001), and an increased risk of cardiovascular events (HR 1.20 [95% CI 1.00-1.44], p = 0.049), with a probability of major cardiovascular events of 13.9% (95% CI 13.3-14.6%) for fertile versus 15.7% (95% CI 14.3-16.9%) for infertile men (p = 0.008), emerged. CONCLUSIONS There is statistical evidence that a diagnosis of male infertility is associated with increased risks of death and incident comorbidities. Owing to the overall high risk of bias, results should be interpreted carefully. PATIENT SUMMARY Male fertility is a proxy of general men's health and as such should be seen as an opportunity to improve preventive strategies for overall men's health beyond the immediate reproductive goals.
Collapse
Affiliation(s)
- Giuseppe Fallara
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy; Department of Urology, IRCCS European Institute of Oncology (IEO), Milan, Italy
| | - Edoardo Pozzi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Federico Belladelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Luca Boeri
- Department of Urology, Foundation IRCCS Ca' Granda - Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Paolo Capogrosso
- Department of Urology and Andrology, Ospedale di Circolo and Macchi Foundation, Varese, Italy
| | - Giovanni Corona
- Medical Department, Endocrinology Unit, Azienda Usl, Maggiore-Bellaria Hospital, Bologna, Italy
| | - Alessia D'Arma
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
47
|
Lin Y, Wang Y, Lai T, Teng J, Lin C, Ke C, Yu I, Lee H, Chan C, Tung C, Conrad DF, O'Bryan MK, Lin Y. Deleterious genetic changes in AGTPBP1 result in teratozoospermia with sperm head and flagella defects. J Cell Mol Med 2024; 28:e18031. [PMID: 37937809 PMCID: PMC10826451 DOI: 10.1111/jcmm.18031] [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: 04/20/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/09/2023] Open
Abstract
Approximately 10%-15% of couples worldwide are infertile, and male factors account for approximately half of these cases. Teratozoospermia is a major cause of male infertility. Although various mutations have been identified in teratozoospermia, these can vary among ethnic groups. In this study, we performed whole-exome sequencing to identify genetic changes potentially causative of teratozoospermia. Out of seven genes identified, one, ATP/GTP Binding Protein 1 (AGTPBP1), was characterized, and three missense changes were identified in two patients (Affected A: p.Glu423Asp and p.Pro631Leu; Affected B: p.Arg811His). In those two cases, severe sperm head and tail defects were observed. Moreover, AGTPBP1 localization showed a fragmented pattern compared to control participants, with specific localization in the neck and annulus regions. Using murine models, we found that AGTPBP1 is localized in the manchette structure, which is essential for sperm structure formation. Additionally, in Agtpbp1-null mice, we observed sperm head and tail defects similar to those in sperm from AGTPBP1-mutated cases, along with abnormal polyglutamylation tubulin and decreasing △-2 tubulin levels. In this study, we established a link between genetic changes in AGTPBP1 and human teratozoospermia for the first time and identified the role of AGTPBP1 in deglutamination, which is crucial for sperm formation.
Collapse
Affiliation(s)
- Yu‐Hua Lin
- Division of Urology, Department of SurgeryCardinal Tien HospitalNew TaipeiTaiwan
- Department of ChemistryFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Ya‐Yun Wang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Tsung‐Hsuan Lai
- Department of Obstetrics and GynecologyCathay General HospitalTaipeiTaiwan
- School of Medicine, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Jih‐Lung Teng
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chi‐Wei Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chih‐Chun Ke
- Department of UrologyEn Chu Kong HospitalNew Taipei CityTaiwan
| | - I‐Shing Yu
- Laboratory Animal CenterCollege of Medicine, National Taiwan UniversityTaipeiTaiwan
| | - Hui‐Ling Lee
- Department of ChemistryFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chying‐Chyuan Chan
- Department of Obstetrics and GynecologyTaipei City Hospital, Zhongxing Branch and Branch for Women and ChildrenTaipeiTaiwan
| | - Chi‐Hua Tung
- Program of Artificial Intelligence & Information SecurityFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Donald F. Conrad
- Division of Genetics, Oregon National Primate Research CenterBeavertonOregonUSA
| | - Moira K. O'Bryan
- School of BioSciences and Bio21 Institute, The University of MelbourneParkvilleVictoriaAustralia
| | - Ying‐Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| |
Collapse
|
48
|
Khan MR, Akbari A, Nicholas TJ, Castillo-Madeen H, Ajmal M, Haq TU, Laan M, Quinlan AR, Ahuja JS, Shah AA, Conrad DF. Genome sequencing of Pakistani families with male infertility identifies deleterious genotypes in SPAG6, CCDC9, TKTL1, TUBA3C, and M1AP. Andrology 2023:10.1111/andr.13570. [PMID: 38073178 PMCID: PMC11163020 DOI: 10.1111/andr.13570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/29/2023] [Accepted: 11/30/2023] [Indexed: 06/12/2024]
Abstract
BACKGROUND There are likely to be hundreds of monogenic forms of human male infertility. Whole genome sequencing (WGS) is the most efficient way to make progress in mapping the causative genetic variants, and ultimately improve clinical management of the disease in each patient. Recruitment of consanguineous families is an effective approach to ascertain the genetic forms of many diseases. OBJECTIVES To apply WGS to large consanguineous families with likely hereditary male infertility and identify potential genetic cases. MATERIALS AND METHODS We recruited seven large families with clinically diagnosed male infertility from rural Pakistan, including five with a history of consanguinity. We generated WGS data on 26 individuals (3-5 per family) and analyzed the resulting data with a computational pipeline to identify potentially causal single nucleotide variants, indels, and copy number variants. RESULTS We identified plausible genetic causes in five of the seven families, including a homozygous 10 kb deletion of exon 2 in a well-established male infertility gene (M1AP), and biallelic missense substitutions (SPAG6, CCDC9, TUBA3C) and an in-frame hemizygous deletion (TKTL1) in genes with emerging relevance. DISCUSSION AND CONCLUSION The rate of genetic findings using the current approach (71%) was much higher than what we recently achieved using whole-exome sequencing (WES) of unrelated singleton cases (20%). Furthermore, we identified a pathogenic single-exon deletion in M1AP that would be undetectable by WES. Screening more families with WGS, especially in underrepresented populations, will further reveal the types of variants underlying male infertility and accelerate the use of genetics in the patient management.
Collapse
Affiliation(s)
- Muhammad Riaz Khan
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Arvand Akbari
- Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Portland, Oregon, USA
| | - Thomas J Nicholas
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Helen Castillo-Madeen
- Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Portland, Oregon, USA
| | - Muhammad Ajmal
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Taqweem Ul Haq
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Maris Laan
- Chair of Human Genetics, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Aaron R Quinlan
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Jasvinder S Ahuja
- Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Portland, Oregon, USA
| | - Aftab Ali Shah
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Donald F Conrad
- Center for Embryonic Cell & Gene Therapy, Oregon Health & Science University, Portland, Oregon, USA
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
49
|
Verpoest W, Okutman Ö, Van Der Kelen A, Sermon K, Viville S. Genetics of infertility: a paradigm shift for medically assisted reproduction. Hum Reprod 2023; 38:2289-2295. [PMID: 37801292 DOI: 10.1093/humrep/dead199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/12/2023] [Indexed: 10/07/2023] Open
Abstract
The field of reproductive genetics has undergone significant advancements with the completion of the Human Genome Project and the development of high-throughput sequencing techniques. This has led to the identification of numerous genes involved in both male and female infertility, revolutionizing the diagnosis and management of infertility patients. Genetic investigations, including karyotyping, specific genetic tests, and high-throughput sequencing, have become essential in determining the genetic causes of infertility. Moreover, the integration of genetics into reproductive medicine has expanded the scope of care to include not only affected individuals or couples but also their family members. Genetic consultations and counselling play a crucial role in identifying potentially affected relatives and offering tailored therapy and the possibility of fertility preservation. Despite the current limited therapeutic options, an increasing understanding of genotype-phenotype correlations in infertility genes holds promise for improved treatment outcomes. The availability of genetic diagnostic tools has reduced the number of idiopathic infertility cases by providing accurate aetiological diagnoses. The transition from research to clinical practice in reproductive genetics requires the establishment of genetic consultations and data warehousing systems to provide up-to-date information on gene-disease relationships. Overall, the integration of genetics into reproductive medicine has brought about a paradigm shift, emphasizing the familial dimension of infertility and offering new possibilities for personalized care and family planning.
Collapse
Affiliation(s)
- Willem Verpoest
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Genetics of Reproduction and Development, Brussels IVF Centre for Reproductive Medicine, Brussels, Belgium
| | - Özlem Okutman
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Hôpital Erasme, Service de Gynécologie-Obstetrique, Clinique de Fertilité, Route de Lennik, Bruxelles, Belgium
| | - Annelore Van Der Kelen
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Genetics of Reproduction and Development, Centre for Medical Genetics, Brussels, Belgium
| | - Karen Sermon
- Vrije Universiteit Brussel (VUB), Faculty of Medicine and Pharmacy, Research Group Genetics of Reproduction and Development, Brussels, Belgium
| | - Stéphane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France
- Laboratoire de Diagnostic Génétique, Unité de Génétique de l'infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
50
|
Zhao J, Ji Z, Meng G, Luo J, Zhang Y, Ou N, Bai H, Tian R, Zhi E, Huang Y, Liu N, He W, Tan Y, Li Z, Yao C, Li P. Identification of a missense variant of MND1 in meiotic arrest and non-obstructive azoospermia. J Hum Genet 2023; 68:729-735. [PMID: 37365320 DOI: 10.1038/s10038-023-01172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023]
Abstract
Meiotic arrest is a common pathologic phenotype of non-obstructive azoospermia (NOA), yet its genetic causes require further investigation. Meiotic nuclear divisions 1 (MND1) has been proved to be indispensable for meiotic recombination in many species. To date, only one variant of MND1 has been reported associated with primary ovarian insufficiency (POI), yet there has been no report of variants in MND1 associated with NOA. Herein, we identified a rare homozygous missense variant (NM_032117:c.G507C:p.W169C) of MND1 in two NOA-affected patients from one Chinese family. Histological analysis and immunohistochemistry demonstrated meiotic arrest at zygotene-like stage in prophase I and lack of spermatozoa in the proband's seminiferous tubules. In silico modeling demonstrated that this variant might cause possible conformational change in the leucine zippers 3 with capping helices (LZ3wCH) domain of MND1-HOP2 complex. Altogether, our study demonstrated that the MND1 variant (c.G507C) is likely responsible for human meiotic arrest and NOA. And our study provides new insights into the genetic etiology of NOA and mechanisms of homologous recombination repair in male meiosis.
Collapse
Affiliation(s)
- Jingpeng Zhao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211100, China
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhiyong Ji
- Department of Reproductive Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Guiquan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China
| | - Jiaqiang Luo
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yuxiang Zhang
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ningjing Ou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211100, China
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haowei Bai
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ruhui Tian
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Erlei Zhi
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yuhua Huang
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Nachuan Liu
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenbin He
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA; National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China
| | - Yueqiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA; National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China
| | - Zheng Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211100, China.
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Chencheng Yao
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Peng Li
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|