1
|
Saez Lancellotti TE, Avena MV, Funes AK, Bernal-López MR, Gómez-Huelgas R, Fornes MW. Exploring the impact of lipid stress on sperm cytoskeleton: insights and prospects. Nat Rev Urol 2025; 22:294-312. [PMID: 39528754 DOI: 10.1038/s41585-024-00952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The decline in male fertility correlates with the global rise in obesity and dyslipidaemia, representing significant public health challenges. High-fat diets induce metabolic alterations, including hypercholesterolaemia, hepatic steatosis and atherosclerosis, with detrimental effects on testicular function. Testicular tissue, critically dependent on lipids for steroidogenesis, is particularly vulnerable to these metabolic disruptions. Excessive lipid accumulation within the testes, including cholesterol, triglycerides and specific fatty acids, disrupts essential sperm production processes such as membrane formation, maturation, energy metabolism and cell signalling. This leads to apoptosis, impaired spermatogenesis, and abnormal sperm morphology and function, ultimately compromising male fertility. During spermiogenesis, round spermatids undergo extensive reorganization, including the formation of the acrosome, manchette and specialized filamentous structures, which are essential for defining the final sperm cell shape. In this Perspective, we examine the impact of high-fat diets on the cytoskeleton of spermatogenic cells and its consequences to identify the mechanisms underlying male infertility associated with dyslipidaemia. Understanding these processes may facilitate the development of therapeutic strategies, such as dietary interventions or natural product supplementation, that aim to address infertility in men with obesity and hypercholesterolaemia. The investigation of cytoskeleton response to lipid stress extends beyond male reproduction, offering insights with broader implications.
Collapse
Affiliation(s)
- Tania E Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina.
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina.
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.
| | - María V Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
2
|
Zheng N, Shen Y, Wang Y, Xiang M, Yu K, Zhang J, Zha X, Duan Z, Wang F, Zhu F, Cao Y. Unraveling the Impact of the PROCA1 Mutation in Male Infertility: Incorporating Whole Exome Sequencing in Teratozoospermia Patients and Analyzing Proca1 Knockout Mice. Reprod Sci 2025; 32:1080-1091. [PMID: 38867036 DOI: 10.1007/s43032-024-01624-6] [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: 12/26/2023] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
In the world, about 15% of couples are infertile, and nearly half of all infertility was caused by men. A large number of genetic mutations are thought to affect spermatogenesis by regulating acrosome formation. Here, we identified three patients harbouring the protein interacting with cyclin A1 (PROCA1) mutation by whole exome sequencing (WES) and Sanger sequencing among patients with predominantly acrosome-deficient teratozoospermia. However, the expression and roles of PROCA1 in infertile men remain unclear. We found that PROCA1 is predominantly expressed in the testis, where it is specifically localized to the acrosome of normal human sperm. Proca1 knockout (KO) mice were subsequently generated using CRISPR-Cas9 technology. However, Proca1 KO adult male mice were fertile, with testis-to-body weight ratios comparable to those of wild-type (WT) mice. Testicular tissue or sperm morphology were not significantly different in Proca1 KO mice compared to WT mice. Expression of the acrosome markers PNA and SP56 in the acrosome was comparable between Proca1 KO and WT mice. In summary, these findings suggested that the PROCA1 mutation identified in humans does not affect acrosome biogenesis in mice.
Collapse
Affiliation(s)
- Na Zheng
- 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, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China
| | - Yiru Shen
- School of Life Science, Anhui Medical University, Hefei, 230022, China
| | - Yu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University), Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China
| | - Mingfei Xiang
- 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, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China
| | - Kexin Yu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University), Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China
| | - Jingjing Zhang
- 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, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, 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, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, 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, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China
| | - Fengsong Wang
- School of Life Science, Anhui Medical University, Hefei, 230022, China.
| | - Fuxi Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University), Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China.
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China.
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, 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, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230032, Anhui, China.
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, 230032, Anhui, China.
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, Anhui, China.
| |
Collapse
|
3
|
Wanjari UR, Gopalakrishnan AV. Exploring the therapeutic effect of melatonin targeting common biomarkers in testicular germ cell tumor, prostate adenocarcinoma, and male infertility: an integrated biology approach. Mamm Genome 2025:10.1007/s00335-025-10119-x. [PMID: 40056207 DOI: 10.1007/s00335-025-10119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
Globally, male infertility (MI) is a major concern. Several other comorbidities related to MI are testicular germ cell tumor (TGCT) and prostate adenocarcinoma (PRAD). This study focuses on finding the common biomarkers among these diseases and their interaction with Melatonin (MLT). The differential expressed genes were retrieved using the GEPIA2 database for TGCT and PRAD, whereas the DISGENET database for MI-related genes. InteractiVenn was performed in response to identify the common genes. The STAG3, RNF212, DDX3Y, DPY19L2, TPCN1, KLK3, GNRH1, DMD, CCDC146, and DNAH1 are found to be involved in all these diseases. The gene ontologies and pathway enrichment analysis were done for these significant genes in response to identifying and accessing the involvement of these genes in other processes. MLT is a neuroendocrine hormone with high therapeutic properties. MLT showed the best binding energy with DDX3Y among all the proteins. Molecular dynamic simulation (MDS) of MLT with DDX3Y was performed and found to be -52.382 ± 13.110 kJ/mol binding energy. The RMSD, RMSF, SASA, RG, H-bond, FEL, PCA, and MM-PBSA analysis confirm the stability and compactness of the DDX3Y-MLT complex. The MDS results indicate that MLT is a promising therapeutic option for enhancing DDX3Y expression, which will support spermatogenesis. Additionally, the hub genes were identified based on MCC parameters from the merged interactive network of common genes in response to finding significant genes that can be a potential biomarker for the diagnosis of diseases.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
He J, Lin X, Tan C, Li Y, Su L, Lin G, Tan YQ, Tu C. Molecular insights into sperm head shaping and its role in human male fertility. Hum Reprod Update 2025:dmaf003. [PMID: 40037590 DOI: 10.1093/humupd/dmaf003] [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: 08/27/2024] [Revised: 12/16/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Sperm head shaping, controlled by the acrosome-acroplaxome-manchette complex, represents a significant morphological change during spermiogenesis and involves numerous proteins expressed in a spatially and temporally specific manner. Defects in sperm head shaping frequently lead to teratozoospermia concomitant with oligozoospermia and asthenozoospermia, but the pathogenic mechanism underlying sperm head shaping, and its role in male infertility, remain poorly understood. OBJECTIVE AND RATIONALE This review aims to summarize the mechanism underlying sperm head shaping, reveal the relationship between gene defects associated with sperm head shaping and male infertility in humans and mice, and explore potential clinical improvements in ICSI treatment. SEARCH METHODS We searched the PubMed database for articles published in English using the keyword 'sperm head shaping' in combination with the following terms: 'acrosome formation', 'proacrosomal vesicles (PAVs)', 'manchette', 'perinuclear theca (PT)', 'chromatin condensation', 'linker of nucleoskeleton and cytoskeleton (LINC) complex', 'histone-to-protamine (HTP) transition', 'male infertility', 'ICSI', and 'artificial oocyte activation (AOA)'. The selected publications until 1 August 2024 were critically summarized, integrated, and thoroughly discussed, and the irrelevant literature were excluded. OUTCOMES A total of 6823 records were retrieved. After careful screening, integrating relevant literature, and excluding articles unrelated to the topic of this review, 240 articles were ultimately included in the analysis. Firstly, we reviewed the important molecular events and structures integral to sperm head shaping, including PAV formation to fusion, acrosome attachment to the nucleus, structure and function of the manchette, PT, chromatin condensation, and HTP transition. Then, we set forth human male infertility associated with sperm head shaping and identified genes related to sperm head shaping resulting in teratozoospermia concomitant with oligozoospermia and asthenozoospermia. Finally, we summarized the outcomes of ICSI in cases of male infertility resulting from mutations in the genes associated with sperm head shaping, as well as the ICSI outcomes through AOA for infertile men with impaired sperm head. WIDER IMPLICATIONS Understanding the molecular mechanisms of sperm head shaping and its relationship with human male infertility holds profound clinical implications, which may contribute to risk prediction, genetic diagnosis, and the potential treatment of human male infertility.
Collapse
Affiliation(s)
- Jiaxin He
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Xinle Lin
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Chen Tan
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yong Li
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Lilan Su
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ge Lin
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Yue-Qiu Tan
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Chaofeng Tu
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| |
Collapse
|
5
|
Barth A, Perry VEA, Hamilton LE, Sutovsky P, Oko R. Bovine Spermatogenesis. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2025; 240:65-136. [PMID: 40272587 DOI: 10.1007/978-3-031-70126-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The intent of this chapter is to provide a morphological foundation in the normal cellular process of bovine gamete development so that abnormalities occurring are recognizable. The knowledge gained here is essential to begin to understand the significance of many of the common bull sperm abnormalities encountered in the clinics. Spermatogenesis is divided into three phases (i. e., Mitosis, Meiosis and Spermiogenesis) all happening in the seminiferous epithelium. The 'Cycle of the Bovine Seminiferous Epithelium' is explained in relation to these phases. Information is provided as to how to identify the stages of the bovine cycle and the steps of spermiogenesis at the histological and ultrastructural levels in preparation to recognize where and when in the cycle a spermatid abnormality arises. Spermiogenesis, the last phase of spermatogenesis, is the most revealing phase to recognize gamete abnormalities as this is where spermatid head and tail differentiation take place and spermatid compartments materialize. The formation of the nucleus, acrosome, manchette, perinuclear theca, axoneme, outer dense fibers, fibrous sheath, connecting piece and mitochondrial sheath occur during this phase and are evaluated. The origins and assembly of a number of essential proteins compartmentalizing the sperm head and tail as well as defects arising during spermiogenesis are reviewed.
Collapse
Affiliation(s)
- Albert Barth
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Viv E A Perry
- Queensland Sperm Morphology Laboratory (QSML), Goondiwindi, QLD, Australia
| | - Lauren E Hamilton
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Science and Department of Obstetrics, Gynecology & Women's Health, University of Missouri, Columbia, MO, USA
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
6
|
Barth A, Perry VEA, Hamilton LE, Sutovsky P, Oko R. Mechanisms of Development of Sperm Defects. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2025; 240:281-296. [PMID: 40272591 DOI: 10.1007/978-3-031-70126-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The intention of this chapter is to provide insights on the possible causes and disruptive mechanisms in play of some commonly occurring bull sperm abnormalities. Consideration is given to mutations of genes, inhibition of enzymes, deficiencies of trace elements, toxins, simulated stress, hormonal changes, hypoosmolarity and cold shock that cause similar disruptive phenotypes in development and structure seen in commonly occurring bull sperm abnormalities. The possible causes and disruptive mechanisms of the following bull sperm defects, arising from the above research, are evaluated: Tail stump, Decapitations, Dag, Dag-like, Short tail, Pseudodoplet, Segmental aplasia of the mitochondrial sheath, Coiled tails, Knobbed and Nuclear vacuolation. In addition, the idea arising from murid research, that mutations affecting sperm head shape most always affect motility, while mutations affecting sperm tail formation rarely affect sperm head shape is considered. Examples of mutations in genes or inhibition of enzymes involved with the early stages of acrosome formation are given that lead to a variety of 'globozoospermic-like' sperm head phenotypes all of which are associated with various degrees of aberrant sperm tail morphologies.
Collapse
Affiliation(s)
- Albert Barth
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Viv E A Perry
- Queensland Sperm Morphology Laboratory (QSML), Goondiwindi, QLD, Australia
| | - Lauren E Hamilton
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Science and Department of Obstetrics, Gynecology & Women's Health, University of Missouri, Columbia, MO, USA
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
7
|
Ji R, Chiozzi RZ, van den Toorn H, Leung M, Zeev-Ben-Mordehai T, Burke ND, Bromfield EG, Reiding KR, Heck AJR. Spatial Organization of the Sperm Cell Glycoproteome. Mol Cell Proteomics 2025; 24:100893. [PMID: 39674511 PMCID: PMC11774830 DOI: 10.1016/j.mcpro.2024.100893] [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: 09/03/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024] Open
Abstract
Sperm cells are terminally differentiated cells that are essential for reproduction in sexually reproducing species. Consistent with their highly specialized function, sperm cells harbor a unique proteome containing many proteins not expressed in somatic cells. In contrast, the post-translational landscape of the sperm proteome remains largely unexplored, limiting our understanding of how modifications such as glycosylation impact sperm function and sperm-egg interactions. Here, we used glycopeptide-centric glycoproteomics to comprehensively characterize protein N-glycosylation in sperm from three mammalian species, revealing clear conservation of glycosylation profiles. We find that glycosylation patterns in sperm proteins are distinct from those in plasma, with as clear distinctive features less sialyation and more paucimannosylation in sperm. Moreover, based on their subcellular location, sperm protein glycosylation varies, with paucimannose species enriched in the acrosomal vesicle, oligomannose species in the sperm head membrane, and complex glycan species in the acrosomal membrane.
Collapse
Affiliation(s)
- Rensong Ji
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Netherlands Proteomic Center, Utrecht, The Netherlands
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Netherlands Proteomic Center, Utrecht, The Netherlands; Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, United Kingdom; Division of Biosciences, University College London Mass Spectrometry Science Technology Platform, University College London, London, United Kingdom
| | - Henk van den Toorn
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Netherlands Proteomic Center, Utrecht, The Netherlands
| | - Miguel Leung
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Tzviya Zeev-Ben-Mordehai
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Nathan D Burke
- School of BioSciences, Faculty of Science, Bio21 Institute, University of Melbourne, Parkville, VIC, Australia; Infertility and Reproduction Research Program, School of Environment and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
| | - Elizabeth G Bromfield
- School of BioSciences, Faculty of Science, Bio21 Institute, University of Melbourne, Parkville, VIC, Australia; Infertility and Reproduction Research Program, School of Environment and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia; Department of Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - Karli R Reiding
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Netherlands Proteomic Center, Utrecht, The Netherlands.
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Netherlands Proteomic Center, Utrecht, The Netherlands.
| |
Collapse
|
8
|
Zhou D, Wu H, Wang L, Wang X, Tang S, Zhou Y, Wang J, Wu B, Tang J, Zhou X, Tian S, Liu S, Lv M, He X, Jin L, Shi H, Zhang F, Cao Y, Liu C. Deficiency of MFSD6L, an acrosome membrane protein, causes oligoasthenoteratozoospermia in humans and mice. J Genet Genomics 2024; 51:1007-1019. [PMID: 38909778 DOI: 10.1016/j.jgg.2024.06.008] [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: 04/18/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024]
Abstract
Oligoasthenoteratozoospermia is an important factor affecting male fertility and has been found to be associated with genetic factors. However, there are still a proportion of oligoasthenoteratozoospermia cases that cannot be explained by known pathogenic genetic variants. Here, we perform genetic analyses and identify bi-allelic loss-of-function variants of MFSD6L from an oligoasthenoteratozoospermia-affected family. Mfsd6l knock-out male mice also present male subfertility with reduced sperm concentration, motility, and deformed acrosomes. Further mechanistic analyses reveal that MFSD6L, as an acrosome membrane protein, plays an important role in the formation of acrosome by interacting with the inner acrosomal membrane protein SPACA1. Moreover, poor embryonic development is consistently observed after intracytoplasmic sperm injection treatment using spermatozoa from the MFSD6L-deficient man and male mice. Collectively, our findings reveal that MFSD6L is required for the anchoring of sperm acrosome and head shaping. The deficiency of MFSD6L affects male fertility and causes oligoasthenoteratozoospermia in humans and mice.
Collapse
Affiliation(s)
- Dapeng Zhou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Lingbo Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Xuemei Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Shuyan Tang
- Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yiling Zhou
- Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Jiaxiong Wang
- State Key Laboratory of Reproductive Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, China; Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, China
| | - Bangguo Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Jianan Tang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Xuehai Zhou
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Shixiong Tian
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Shuang Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| | - Huijuan Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Feng Zhang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China.
| | - Chunyu Liu
- Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
9
|
Miyata H, Shimada K, Kaneda Y, Ikawa M. Development of functional spermatozoa in mammalian spermiogenesis. Development 2024; 151:dev202838. [PMID: 39036999 DOI: 10.1242/dev.202838] [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: 07/23/2024]
Abstract
Infertility is a global health problem affecting one in six couples, with 50% of cases attributed to male infertility. Spermatozoa are male gametes, specialized cells that can be divided into two parts: the head and the flagellum. The head contains a vesicle called the acrosome that undergoes exocytosis and the flagellum is a motility apparatus that propels the spermatozoa forward and can be divided into two components, axonemes and accessory structures. For spermatozoa to fertilize oocytes, the acrosome and flagellum must be formed correctly. In this Review, we describe comprehensively how functional spermatozoa develop in mammals during spermiogenesis, including the formation of acrosomes, axonemes and accessory structures by focusing on analyses of mouse models.
Collapse
Affiliation(s)
- Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Kaneda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
10
|
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
|
11
|
Wang Y, Huang X, Sun G, Chen J, Wu B, Luo J, Tang S, Dai P, Zhang F, Li J, Wang L. Coiled-coil domain-containing 38 is required for acrosome biogenesis and fibrous sheath assembly in mice. J Genet Genomics 2024; 51:407-418. [PMID: 37709195 DOI: 10.1016/j.jgg.2023.09.002] [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: 05/23/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023]
Abstract
During spermiogenesis, haploid spermatids undergo dramatic morphological changes to form slender sperm flagella and cap-like acrosomes, which are required for successful fertilization. Severe deformities in flagella cause a male infertility syndrome, multiple morphological abnormalities of the flagella (MMAF), while acrosomal hypoplasia in some cases leads to sub-optimal embryonic developmental potential. However, evidence regarding the occurrence of acrosomal hypoplasia in MMAF is limited. Here, we report the generation of base-edited mice knocked out for coiled-coil domain-containing 38 (Ccdc38) via inducing a nonsense mutation and find that the males are infertile. The Ccdc38-KO sperm display acrosomal hypoplasia and typical MMAF phenotypes. We find that the acrosomal membrane is loosely anchored to the nucleus and fibrous sheaths are disorganized in Ccdc38-KO sperm. Further analyses reveal that Ccdc38 knockout causes a decreased level of TEKT3, a protein associated with acrosome biogenesis, in testes and an aberrant distribution of TEKT3 in sperm. We finally show that intracytoplasmic sperm injection overcomes Ccdc38-related infertility. Our study thus reveals a previously unknown role for CCDC38 in acrosome biogenesis and provides additional evidence for the occurrence of acrosomal hypoplasia in MMAF.
Collapse
Affiliation(s)
- Yaling Wang
- 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
| | - 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
| | - Guoying Sun
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Jingwen Chen
- 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
| | - Bangguo Wu
- 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
| | - Jiahui Luo
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Shuyan Tang
- Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, 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
- 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
- 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
|
12
|
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: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
13
|
Fujihara Y, Miyata H, Abbasi F, Larasati T, Nozawa K, Yu Z, Ikawa M, Matzuk MM. Tex46 knockout male mice are sterile secondary to sperm head malformations and failure to penetrate through the zona pellucida. PNAS NEXUS 2024; 3:pgae108. [PMID: 38516277 PMCID: PMC10957234 DOI: 10.1093/pnasnexus/pgae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
Each year, infertility affects 15% of couples worldwide, with 50% of cases attributed to men. It is assumed that sperm head shape is important for sperm-zona pellucida (ZP) penetration but research has yet to elucidate why. We generated testis expressed 46 (Tex46) knockout mice to investigate the essential roles of TEX46 in mammalian reproduction. We used RT-PCR to demonstrate that Tex46 was expressed exclusively in the male reproductive tract in mice and humans. We created Tex46-/- mice using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system and analyzed their fertility. Tex46 null spermatozoa underwent further evaluation using computer-assisted sperm analysis, light microscopy, and ultrastructural microscopy. We used immunoblot analysis to elucidate relationships between TEX46 and other acrosome biogenesis-related proteins. Mouse and human TEX46 are testis-enriched and encode a transmembrane protein which is conserved from amphibians to mammals. Loss of the mouse TEX46 protein causes male sterility primarily due to abnormal sperm head formation and secondary effects on sperm motility. Tex46 null spermatozoa morphologically lack the typical hooked sperm head appearance and fail to penetrate through the ZP. Electron microscopy of the testicular germ cells reveals malformation of the acrosomal cap, with misshapen sperm head tips and the appearance of a gap between the acrosome head and the nucleus. TEX46 is essential for sperm head formation, sperm penetration through the ZP, and male fertility in mice, and is a putative contraceptive target in men.
Collapse
Affiliation(s)
- Yoshitaka Fujihara
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Suita, Osaka 564-8565, Japan
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ferheen Abbasi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Tamara Larasati
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kaori Nozawa
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Suita, Osaka 564-8565, Japan
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhifeng Yu
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
14
|
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
|
15
|
Clark AC, Edison R, Esvelt K, Kamau S, Dutoit L, Champer J, Champer SE, Messer PW, Alexander A, Gemmell NJ. A framework for identifying fertility gene targets for mammalian pest control. Mol Ecol Resour 2024; 24:e13901. [PMID: 38009398 PMCID: PMC10860713 DOI: 10.1111/1755-0998.13901] [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: 06/23/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023]
Abstract
Fertility-targeted gene drives have been proposed as an ethical genetic approach for managing wild populations of vertebrate pests for public health and conservation benefit. This manuscript introduces a framework to identify and evaluate target gene suitability based on biological gene function, gene expression and results from mouse knockout models. This framework identified 16 genes essential for male fertility and 12 genes important for female fertility that may be feasible targets for mammalian gene drives and other non-drive genetic pest control technology. Further, a comparative genomics analysis demonstrates the conservation of the identified genes across several globally significant invasive mammals. In addition to providing important considerations for identifying candidate genes, our framework and the genes identified in this study may have utility in developing additional pest control tools such as wildlife contraceptives.
Collapse
Affiliation(s)
- Anna C Clark
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Rey Edison
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Kevin Esvelt
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Sebastian Kamau
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Ludovic Dutoit
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin 9016, New Zealand
| | - Jackson Champer
- Center for Bioinformatics, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Samuel E Champer
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Philipp W Messer
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Alana Alexander
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
| | - Neil J Gemmell
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
| |
Collapse
|
16
|
Wei YL, Fan XJ, Lin XC, Lin AZ, She ZY, Wang XR. Kinesin-14 KIFC1 promotes acrosome formation and chromatin maturation during mouse spermiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119555. [PMID: 37524262 DOI: 10.1016/j.bbamcr.2023.119555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
KIFC1, a member of kinesin-14 subfamily motors, is essential for meiotic cell division and acrosome formation during spermatogenesis. However, the functions of KIFC1 in the formation and maintenance of the acrosome in male germ cells remain to be elucidated. In this study, we report the structural deformities of acrosomes in the in vivo KIFC1 inhibition mouse models. The proacrosomal vesicles diffuse into the cytoplasm and form atypical acrosomal granules. This phenotype is consistent with globozoospermia patients and probably results from the failure of the Golgi-derived vesicle trafficking and actin filament organization. Moreover, the multinucleated and undifferentiated spermatogenic cells in the epidydimal lumen after KIFC1 inhibition reveal the specific roles of KIFC1 in regulating post-meiotic maturation. Overall, our results uncover KIFC1 as an essential regulator in the trafficking, fusion and maturation of acrosomal vesicles during spermiogenesis.
Collapse
Affiliation(s)
- Ya-Lan Wei
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian 350013, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Xiao-Jing Fan
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian 350013, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Xin-Chen Lin
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian 350013, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Ai-Zhu Lin
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian 350013, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| | - Xin-Rui Wang
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian 350013, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350122, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian 350001, China.
| |
Collapse
|
17
|
Bilekova S, Garcia-Colomer B, Cebrian-Serrano A, Schirge S, Krey K, Sterr M, Kurth T, Hauck SM, Lickert H. Inceptor facilitates acrosomal vesicle formation in spermatids and is required for male fertility. Front Cell Dev Biol 2023; 11:1240039. [PMID: 37691832 PMCID: PMC10483240 DOI: 10.3389/fcell.2023.1240039] [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: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Spermatogenesis is a crucial biological process that enables the production of functional sperm, allowing for successful reproduction. Proper germ cell differentiation and maturation require tight regulation of hormonal signals, cellular signaling pathways, and cell biological processes. The acrosome is a lysosome-related organelle at the anterior of the sperm head that contains enzymes and receptors essential for egg-sperm recognition and fusion. Even though several factors crucial for acrosome biogenesis have been discovered, the precise molecular mechanism of pro-acrosomal vesicle formation and fusion is not yet known. In this study, we investigated the role of the insulin inhibitory receptor (inceptor) in acrosome formation. Inceptor is a single-pass transmembrane protein with similarities to mannose-6-phosphate receptors (M6PR). Inceptor knockout male mice are infertile due to malformations in the acrosome and defects in the nuclear shape of spermatozoa. We show that inceptor is expressed in early spermatids and mainly localizes to vesicles between the Golgi apparatus and acrosome. Here we show that inceptor is an essential factor in the intracellular transport of trans-Golgi network-derived vesicles which deliver acrosomal cargo in maturing spermatids. The absence of inceptor results in vesicle-fusion defects, acrosomal malformation, and male infertility. These findings support our hypothesis of inceptor as a universal lysosomal or lysosome-related organelle sorting receptor expressed in several secretory tissues.
Collapse
Affiliation(s)
- Sara Bilekova
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Balma Garcia-Colomer
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Helmholtz Center Munich, Institute for Diabetes and Obesity, Neuherberg, Germany
| | - Alberto Cebrian-Serrano
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Helmholtz Center Munich, Institute for Diabetes and Obesity, Neuherberg, Germany
| | - Silvia Schirge
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Krey
- School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Michael Sterr
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, Core Facility Electron Microscopy and Histology, Dresden University of Technology, Dresden, Germany
| | - Stefanie M. Hauck
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Heiko Lickert
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
18
|
Fujihara Y, Kobayashi K, Abbasi F, Endo T, Yu Z, Ikawa M, Matzuk MM. PDCL2 is essential for sperm acrosome formation and male fertility in mice. Andrology 2023; 11:789-798. [PMID: 36278277 PMCID: PMC10123174 DOI: 10.1111/andr.13329] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Each year, infertility affects 15% of couples worldwide, with 50% of cases attributed to men. Globozoospermia is an uncommon cause of male factor infertility, characterized by defects in sperm acrosome formation, leading to round-headed spermatozoa. OBJECTIVE We generated Pdcl2 knockout mice to investigate the essential roles of PDCL2 in mammalian reproduction. MATERIALS AND METHODS We used reverse transcription-polymerase chain reaction to demonstrate that PDCL2 was expressed exclusively in the male reproductive tract in mice and humans. We created Pdcl2 knockout mice using the CRISPR-Cas9 system and analyzed their fertility. Pdcl2 null spermatozoa underwent further evaluation using computer-assisted sperm analysis, light microscopy, and ultrastructural microscopy. We used immunoblot analysis and immunofluorescence to elucidate relationships between PDCL2 and other acrosomal proteins. RESULTS The PDC family is highly conserved in eukaryotes. Mouse and human PDCL2 are testis enriched and localized to the testicular endoplasmic reticulum. Loss of the protein causes sterility because of abnormal acrosome biogenesis during spermiogenesis and immotility. Furthermore, Pdcl2 null spermatozoa have rounded heads, similar to globozoospermia in humans. Observation of the knockout testis shows a lack of acrosomal cap formation, aberrant localization of mitochondria in the sperm head, and misshapen nuclei. CONCLUSION PDCL2 is essential for sperm acrosome development and male fertility in mice and is a putative contraceptive target in men.
Collapse
Affiliation(s)
- Yoshitaka Fujihara
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Osaka 564-8565, Japan
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ferheen Abbasi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- School of Medicine, University of California, Davis, Sacramento, CA 95817
| | - Tsutomu Endo
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Experimental Animals, Research Facility Cluster, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Zhifeng Yu
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Martin M. Matzuk
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
19
|
Clark AC, Alexander A, Edison R, Esvelt K, Kamau S, Dutoit L, Champer J, Champer SE, Messer PW, Gemmell NJ. A framework for identifying fertility gene targets for mammalian pest control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542751. [PMID: 37398071 PMCID: PMC10312551 DOI: 10.1101/2023.05.30.542751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Fertility-targeted gene drives have been proposed as an ethical genetic approach for managing wild populations of vertebrate pests for public health and conservation benefit.This manuscript introduces a framework to identify and evaluate target gene suitability based on biological gene function, gene expression, and results from mouse knockout models.This framework identified 16 genes essential for male fertility and 12 genes important for female fertility that may be feasible targets for mammalian gene drives and other non-drive genetic pest control technology. Further, a comparative genomics analysis demonstrates the conservation of the identified genes across several globally significant invasive mammals.In addition to providing important considerations for identifying candidate genes, our framework and the genes identified in this study may have utility in developing additional pest control tools such as wildlife contraceptives.
Collapse
Affiliation(s)
- Anna C Clark
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Alana Alexander
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
| | - Rey Edison
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Kevin Esvelt
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Sebastian Kamau
- Media Laboratory, Massachusetts Institute of Technology, 75 Amherst St, Cambridge, United States
| | - Ludovic Dutoit
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin 9016, New Zealand
| | - Jackson Champer
- Center for Bioinformatics, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Samuel E Champer
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Philipp W Messer
- Department of Computational Biology, Cornell University, 102 Tower Rd, Ithaca, NY 14853, United States
| | - Neil J Gemmell
- Department of Anatomy, School of Biomedical Sciences, University of Otago, 270 Great King Street, Central Dunedin, Dunedin 9016, New Zealand
| |
Collapse
|
20
|
Miyazaki MA, Guilharducci RL, Intasqui P, Bertolla RP. Mapping the human sperm proteome - novel insights into reproductive research. Expert Rev Proteomics 2023; 20:19-45. [PMID: 37140161 DOI: 10.1080/14789450.2023.2210764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
INTRODUCTION Spermatozoa are highly specialized cells with unique morphology. In addition, spermatozoa lose a considerable amount of cytoplasm during spermiogenesis, when they also compact their DNA, resulting in a transcriptionally quiescent cell. Throughout the male reproductive tract, sperm will acquire proteins that enable them to interact with the female reproductive tract. After ejaculation, proteins undergo post-translational modifications for sperm to capacitate, hyperactivate and fertilize the oocyte. Many proteins have been identified as predictors of male infertility, and also investigated in diseases that compromise reproductive potential. AREAS COVERED In this review we proposed to summarize the recent findings about the sperm proteome and how they affect sperm structure, function, and fertility. A literature search was performed using PubMed and Google Scholar databases within the past 5 years until August 2022. EXPERT OPINION Sperm function depends on protein abundance, conformation, and PTMs; understanding the sperm proteome may help to identify pathways essential to fertility, even making it possible to unravel the mechanisms involved in idiopathic infertility. In addition, proteomics evaluation offers knowledge regarding alterations that compromise the male reproductive potential.
Collapse
Affiliation(s)
- Mika Alexia Miyazaki
- Department of Surgery, Division of Urology, Human Reproduction Section, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Raquel Lozano Guilharducci
- Department of Surgery, Division of Urology, Human Reproduction Section, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paula Intasqui
- Department of Surgery, Division of Urology, Human Reproduction Section, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ricardo Pimenta Bertolla
- Department of Surgery, Division of Urology, Human Reproduction Section, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Zheng R, Wang Y, Li Y, Guo J, Wen Y, Jiang C, Yang Y, Shen Y. FSIP2 plays a role in the acrosome development during spermiogenesis. J Med Genet 2023; 60:254-264. [PMID: 35654582 DOI: 10.1136/jmedgenet-2021-108406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/11/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Loss-of-function mutations in FSIP2 result in multiple morphological abnormalities of the flagella in humans and mice. Intriguingly, a recent study found that FSIP2 might regulate the expression of acrosomal proteins, indicating that Fsip2 might be involved in acrosome development in mice. However, whether FSIP2 also function in acrosome biogenesis in humans is largely unknown, and the underlying mechanism of which is unexplored. OBJECTIVE Our objective was to reveal potential function of FSIP2 in regulating sperm acrosome formation. METHODS We performed whole exome sequencing on four asthenoteratozoospermic patients. Western blot analysis and immunofluorescence staining were conducted to assess the protein expression of FSIP2. Proteomics approach, liquid chromatography-tandem mass spectrometry and co-immunoprecipitation were implemented to clarify the molecules in acrosome biogenesis regulated by FSIP2. RESULTS Biallelic FSIP2 variants were identified in four asthenoteratozoospermic individuals. The protein expression of MUT-FSIP2 was sharply decreased or absent in vitro or in vivo. Interestingly, aside from the sperm flagellar defects, the acrosomal hypoplasia was detected in numerous sperm from the four patients. FSIP2 co-localised with peanut agglutinin in the acrosome during spermatogenesis. Moreover, FSIP2 interacted with proteins (DPY19L2, SPACA1, HSP90B1, KIAA1210, HSPA2 and CLTC) involved in acrosome biogenesis. In addition, spermatozoa from patients carrying FSIP2 mutations showed downregulated expression of DPY19L2, ZPBP, SPACA1, CCDC62, CCIN, SPINK2 and CSNK2A2. CONCLUSION Our findings unveil that FSIP2 might involve in sperm acrosome development, and consequently, its mutations might contribute to globozoospermia or acrosomal aplasia. We meanwhile first uncover the potential molecular mechanism of FSIP2 regulating acrosome biogenesis.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Wang
- 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, Sichuan, China
| | - Yaqian Li
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Juncen Guo
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yuting Wen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Chuan Jiang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 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, Sichuan, China
| | - Ying Shen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Dai J, Chen Y, Li Q, Zhang T, Zhou Q, Gong F, Lu G, Zheng W, Lin G. Pathogenic variant in ACTL7A causes severe teratozoospermia characterized by bubble-shaped acrosomes and male infertility. Mol Hum Reprod 2022; 28:6648105. [PMID: 35863052 DOI: 10.1093/molehr/gaac028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Teratozoospermia is a common factor associated with male infertility. However, teratozoospermia characterized by bubble-shaped acrosomes (BSAs) has not yet been identified in men and the causative genes are unknown. The present study is of a patient with severe teratozoospermia characterized by BSA and carrying a variant (c.1204G>A, p. Gly402Ser) of actin-like 7A (ACTL7A). For further verification, we generated an Actl7a-mutated mouse model (p.Gly407Ser) carrying an equivalent variant to that in the patient. We found that homozygous Actl7a-mutated (Actl7aMut/Mut) male mice were sterile, and all their sperm showed acrosomal abnormalities. We detected, by transmission electron microscopy, that during acrosomal biogenesis the acrosome detaches from the nuclear membrane in Actl7aMut/Mut mice. Furthermore, mutant ACTL7A failed to attach to the acroplaxome and was discharged by cytoplasmic droplets, which led to the absence of ACTL7A in epididymal spermatozoa in mice. The mutant sperm failed to activate the oocyte, and sperm-borne oocyte activation factor PLCζ discharge accompanied by ACTL7A was observed, leading to total fertilization failure (TFF). Immunoprecipitation followed by liquid chromatography-mass spectrometry showed that several differentially expressed proteins participate in acrosome assembly and actin filament organization. Furthermore, assisted oocyte activation by calcium ionophore exposure successfully overcame TFF in the couple with an ACTL7A pathogenic variant. Our study defined a novel phenotype of an acrosomal abnormality characterized by BSA, revealed the underlying mechanism of a pathogenic variant in ACTL7A, and provided a genetic marker and potential therapeutic option for male infertility.
Collapse
Affiliation(s)
- Jing Dai
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, ChangSha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Yongzhe Chen
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, ChangSha, 410078, China
| | - Qi Li
- Xiangya Hospital Central South University, ChangSha, 410008, China
| | - Tianlei Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Qinwei Zhou
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, ChangSha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Guangxiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Wei Zheng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, ChangSha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, ChangSha, 410078, China
| |
Collapse
|
23
|
The perinuclear theca protein Calicin helps shape the sperm head and maintain the nuclear structure in mice. Cell Rep 2022; 40:111049. [PMID: 35793634 DOI: 10.1016/j.celrep.2022.111049] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/03/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The perinuclear theca (PT) is a cytoskeletal element encapsulating the sperm nucleus; however, our understanding of the physiological roles of PT in sperm is very limited. We show that Calicin interacts with itself and many other PT components, indicating it may serve as an organizing center of the PT assembly. Calicin is detectable first when surrounding the acrosome, then detected around the entire nucleus, and finally translocated to the postacrosomal region of spermatid heads. Intriguingly, loss of Calicin specifically causes surface subsidence of sperm heads in the nuclear condensation stage. Calicin interacts with inner acrosomal membrane (IAM) protein Spaca1 and nuclear envelope (NE) components to form an "IAM-PT-NE" structure. Intriguingly, Ccin-knockout sperm also exhibit DNA damage and failure of fertilization. Our study provides solid animal evidence to suggest that the PT encapsulating sperm nucleus helps shape the sperm head and maintain the nuclear structure.
Collapse
|
24
|
Zhang XZ, Wei LL, Zhang XH, Jin HJ, Chen SR. Loss of perinuclear theca ACTRT1 causes acrosome detachment and severe male subfertility in mice. Development 2022; 149:275523. [DOI: 10.1242/dev.200489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/12/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The perinuclear theca (PT) is a cytoskeletal element encapsulating the sperm nucleus; however, the physiological roles of the PT in sperm are largely uncertain. Here, we reveal that ACTRT1, ACTRT2, ACTL7A and ACTL9 proteins interact to form a multimeric complex and localize to the subacrosomal region of spermatids. Furthermore, we engineered Actrt1-knockout (KO) mice to define the functions of ACTRT1. Despite normal sperm count and motility, Actrt1-KO males were severely subfertile owing to a deficiency in fertilization. Loss of ACTRT1 caused a high incidence of malformed heads and detachment of acrosomes from sperm nuclei, caused by loosened acroplaxome structure during spermiogenesis. Furthermore, Actrt1-KO sperm showed reduced ACTL7A and PLCζ protein content as a potential cause of fertilization defects. Moreover, we reveal that ACTRT1 anchors developing acrosomes to the nucleus, likely by interacting with the inner acrosomal membrane protein SPACA1 and the nuclear envelope proteins PARP11 and SPATA46. Loss of ACTRT1 weakened the interaction between ACTL7A and SPACA1. Our study and recent findings of ACTL7A/ACTL9-deficient sperm together reveal that the sperm PT-specific ARP complex mediates the acrosome-nucleus connection.
Collapse
Affiliation(s)
- Xiao-Zhen Zhang
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Lin-Lin Wei
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Xiao-Hui Zhang
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Hui-Juan Jin
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| | - Su-Ren Chen
- Ministry of Education, Department of Biology, College of Life Sciences Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
- Beijing Normal University Key Laboratory of Cell Proliferation and Regulation Biology , , , 100875 Beijing , China
| |
Collapse
|
25
|
Okada Y. Sperm chromatin condensation: epigenetic mechanisms to compact the genome and spatiotemporal regulation from inside and outside the nucleus. Gene 2022; 97:41-53. [PMID: 35491100 DOI: 10.1266/ggs.21-00065] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Sperm chromatin condensation is a critical step in mammalian spermatogenesis to protect the paternal DNA from external damaging factors and to acquire fertility. During chromatin condensation, various events proceed in a chronological order, independently or in sequence, interacting with each other both inside and outside the nucleus to support the dramatic chromatin changes. Among these events, histone-protamine replacement, which is concomitant with acrosome biogenesis and cytoskeletal alteration, is the most critical step associated with nuclear elongation. Failures of not only intranuclear events but also extra-nuclear events severely affect sperm shape and chromatin state and are subsequently linked to infertility. This review focuses on nuclear and non-nuclear factors that affect sperm chromatin condensation and its effects, and further discusses the possible utility of sperm chromatin for clinical applications.
Collapse
Affiliation(s)
- Yuki Okada
- Laboratory of Pathology and Development, Institute for Quantitative Biosciences, The University of Tokyo
| |
Collapse
|
26
|
Wang HQ, Wang T, Gao F, Ren WZ. Application of CRISPR/Cas Technology in Spermatogenesis Research and Male Infertility Treatment. Genes (Basel) 2022; 13:genes13061000. [PMID: 35741761 PMCID: PMC9223233 DOI: 10.3390/genes13061000] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
As the basis of animal reproductive activity, normal spermatogenesis directly determines the efficiency of livestock production. An in-depth understanding of spermatogenesis will greatly facilitate animal breeding efforts and male infertility treatment. With the continuous development and application of gene editing technologies, they have become valuable tools to study the mechanism of spermatogenesis. Gene editing technologies have provided us with a better understanding of the functions and potential mechanisms of action of factors that regulate spermatogenesis. This review summarizes the applications of gene editing technologies, especially CRISPR/Cas9, in deepening our understanding of the function of spermatogenesis-related genes and disease treatment. The problems of gene editing technologies in the field of spermatogenesis research are also discussed.
Collapse
|
27
|
Ernandez J, Gu C, Kathrins M. Awareness and reporting of globozoospermia among in vitro fertilization and andrology laboratories: A national survey. Andrologia 2022; 54:e14474. [PMID: 35587120 DOI: 10.1111/and.14474] [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: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022] Open
Abstract
Globozoospermia (GZ) is a rare disorder found in less than 0.1% of infertile men in which spermatozoa lack acrosomes necessary for penetration of an oocyte. While methods have been demonstrated to allow globozoospermic men to achieve a viable pregnancy with their partner, the Wold Health Organization considers identifying and reporting GZ on semen analysis to be 'important'. Our study aims to determine if and to what extent in vitro fertilization (IVF) laboratories nationwide recognize and report GZ on semen analysis reports. We constructed an IRB-approved survey sent nationwide to IVF and andrology clinic laboratory directors listed by the Society for Assisted Reproductive Technology and/or the American Society for Reproductive Medicine. Results from the survey were de-identified for analysis. A total of 490 surveys were sent with a response rate of 10% (n = 51). Most respondents (66%) practiced in a private, rather than academic, setting. A majority of respondents were confident in their technicians' knowledge of GZ (86%) and ability to identify it on a sample (94%). However, only half of respondents noted a space to report the concern for GZ to the ordering physician, and 25% of respondents did not feel their clinic was able to identify patients where there is a concern for GZ. Similarly, 84% of respondents did not report a percent of acrosome-deficient sperm. Less than half of respondents reported that their clinic has previously diagnosed GZ. Though the majority of respondents felt that their laboratory technicians would be able to identify GZ, a significant minority felt that their clinic did not have means to be able to report concern for GZ. This may be due to the absence of a proper channel to report a concern for GZ, a lack of knowledge about the condition, or failure to distinguish GZ from a broader reported percentage of morphologically abnormal sperm. Given evidence that the diagnosis of GZ may be under-reported in the United States, there should be a national standard for laboratory technicians to be trained to recognize GZ and be able to report their suspicion to the ordering clinician.
Collapse
Affiliation(s)
- John Ernandez
- Division of Urology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Catherine Gu
- Division of Urology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Martin Kathrins
- Division of Urology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Zhou L, Yu Z, Xia Y, Cheng S, Gao J, Sun W, Jiang X, Zhang J, Mao L, Qin X, Zou Z, Qiu J, Chen C. Repression of autophagy leads to acrosome biogenesis disruption caused by a sub-chronic oral administration of polystyrene nanoparticles. ENVIRONMENT INTERNATIONAL 2022; 163:107220. [PMID: 35381522 DOI: 10.1016/j.envint.2022.107220] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
As a new widespread contaminant, nanoplastics (NPs) pose a potential risk to human health. Nevertheless, the adverse effects of NPs on the male reproductive system are poorly understood. In this study, we aimed to determine the effects of polystyrene nanoplastics (PS-NPs) (50 nm) on sperm quality, with a focus on the acrosome defects. After 35 days of intragastric administration, sperm quality was decreased and testicular structures were impaired in mice exposed to PS-NPs in both the medium (1.0 mg/kg) and high dose (10 mg/kg) groups. No significant changes were observed in the low dose (0.2 mg/kg) group. Meanwhile, acrosome parameters including acrosome integrity and acrosome reaction were decreased after the administration of PS-NPs. These findings were consistent with the disruption of acrosome biogenesis, as identified by the changed testicular ultrastructure. Additionally, the findings were further validated using seven marker genes (Gba2, Pick1, Gopc, Hrb, Zpbp1, Spaca1 and Dpy19l2) essential for acrosome formation, which showed that two of these genes (Gopc and Dpy19l2) were significantly down-regulated. Moreover, repressed autophagy was observed in the testes of PS-NPs-exposed mice based on autophagy-related protein expression. This phenomenon was further verified in GC-2spd cells treated with PS-NPs (50 μg/mL, 100 μg/mL, 200 μg/mL for 24 h). The potential role of autophagy in such acrosome defects was explored by using the autophagy inhibitor 3-methyladenine (3-MA), autophagy activator rapamycin or beclin-1 siRNA. The results showed that Golgi-associated vesicle disorganization was exacerbated with the 3-MA and beclin-1 siRNA pretreatments, but decreased with the rapamycin pretreatment, and the expression of GOPC and DPY19L2 was also altered. These results indicated that autophagy might be involved in the PS-NPs-induced acrosome lesions based on the regulation of two key acrosome-formation proteins, GOPC and DPY19L2. Altogether, our results will provide new insights into the PS-NPs-induced male reproductive impairment.
Collapse
Affiliation(s)
- Lixiao Zhou
- Department of Health Laboratory Technology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Ziying Yu
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jieying Gao
- Department of Health Laboratory Technology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Wei Sun
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China; Molecular Biology Laboratory of Respiratory Diseases, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lejiao Mao
- Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China; Molecular Biology Laboratory of Respiratory Diseases, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China; Molecular Biology Laboratory of Respiratory Diseases, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
29
|
Genome-wide compound heterozygote analysis highlights DPY19L2 alleles in a non-consanguineous Spanish family with a complete form of globozoospermia. Reprod Biomed Online 2022; 45:332-340. [DOI: 10.1016/j.rbmo.2022.03.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022]
|
30
|
Teves ME, Roldan ERS. Sperm bauplan and function and underlying processes of sperm formation and selection. Physiol Rev 2022; 102:7-60. [PMID: 33880962 PMCID: PMC8812575 DOI: 10.1152/physrev.00009.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
The spermatozoon is a highly differentiated and polarized cell, with two main structures: the head, containing a haploid nucleus and the acrosomal exocytotic granule, and the flagellum, which generates energy and propels the cell; both structures are connected by the neck. The sperm's main aim is to participate in fertilization, thus activating development. Despite this common bauplan and function, there is an enormous diversity in structure and performance of sperm cells. For example, mammalian spermatozoa may exhibit several head patterns and overall sperm lengths ranging from ∼30 to 350 µm. Mechanisms of transport in the female tract, preparation for fertilization, and recognition of and interaction with the oocyte also show considerable variation. There has been much interest in understanding the origin of this diversity, both in evolutionary terms and in relation to mechanisms underlying sperm differentiation in the testis. Here, relationships between sperm bauplan and function are examined at two levels: first, by analyzing the selective forces that drive changes in sperm structure and physiology to understand the adaptive values of this variation and impact on male reproductive success and second, by examining cellular and molecular mechanisms of sperm formation in the testis that may explain how differentiation can give rise to such a wide array of sperm forms and functions.
Collapse
Affiliation(s)
- Maria Eugenia Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia
| | - Eduardo R S Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales (CSIC), Madrid, Spain
| |
Collapse
|
31
|
Yogo K. Molecular basis of the morphogenesis of sperm head and tail in mice. Reprod Med Biol 2022; 21:e12466. [PMID: 35619659 PMCID: PMC9126569 DOI: 10.1002/rmb2.12466] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022] Open
Abstract
Background The spermatozoon has a complex molecular apparatus necessary for fertilization in its head and flagellum. Recently, numerous genes that are needed to construct the molecular apparatus of spermatozoa have been identified through the analysis of genetically modified mice. Methods Based on the literature information, the molecular basis of the morphogenesis of sperm heads and flagella in mice was summarized. Main findings (Results) The molecular mechanisms of vesicular trafficking and intraflagellar transport in acrosome and flagellum formation were listed. With the development of cryo‐electron tomography and mass spectrometry techniques, the details of the axonemal structure are becoming clearer. The fine structure and the proteins needed to form the central apparatus, outer and inner dynein arms, nexin‐dynein regulatory complex, and radial spokes were described. The important components of the formation of the mitochondrial sheath, fibrous sheath, outer dense fiber, and the annulus were also described. The similarities and differences between sperm flagella and Chlamydomonas flagella/somatic cell cilia were also discussed. Conclusion The molecular mechanism of formation of the sperm head and flagellum has been clarified using the mouse as a model. These studies will help to better understand the diversity of sperm morphology and the causes of male infertility.
Collapse
Affiliation(s)
- Keiichiro Yogo
- Department of Applied Life Sciences Faculty of Agriculture Shizuoka University Shizuoka Japan
| |
Collapse
|
32
|
Cazin C, Neirijnck Y, Loeuillet C, Wehrli L, Kühne F, Lordey I, Mustapha SFB, Bouker A, Zouari R, Thierry-Mieg N, Nef S, Arnoult C, Ray PF, Kherraf ZE. Combined Use of Whole Exome Sequencing and CRISPR/Cas9 to Study the Etiology of Non-Obstructive Azoospermia: Demonstration of the Dispensable Role of the Testis-Specific Genes C1orf185 and CCT6B. Cells 2021; 11:cells11010118. [PMID: 35011680 PMCID: PMC8750304 DOI: 10.3390/cells11010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
The genetic landscape of male infertility is highly complex. It is estimated that at least 4000 genes are involved in human spermatogenesis, but only few have so far been extensively studied. In this study, we investigated by whole exome sequencing two cases of idiopathic non-obstructive azoospermia (NOA) due to severe hypospermatogenesis. After variant filtering and prioritizing, we retained for each patient a homozygous loss-of-function (LoF) variant in a testis-specific gene, C1orf185 (c.250C>T; p.Gln84Ter) and CCT6B (c.615-2A>G), respectively. Both variants are rare according to the gnomAD database and absent from our local control cohort (n = 445). To verify the implication of these candidate genes in NOA, we used the CRISPR/Cas9 system to invalidate the mouse orthologs 4930522H14Rik and Cct6b and produced two knockout (KO) mouse lines. Sperm and testis parameters of homozygous KO adult male mice were analyzed and compared with those of wild-type animals. We showed that homozygous KO males were fertile and displayed normal sperm parameters and a functional spermatogenesis. Overall, these results demonstrate that not all genes highly and specifically expressed in the testes are essential for spermatogenesis, and in particular, we conclude that bi-allelic variants of C1orf185 and CCT6B are most likely not to be involved in NOA and male fertility.
Collapse
Affiliation(s)
- Caroline Cazin
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France; (C.C.); (C.L.); (C.A.); (P.F.R.)
- UM GI-DPI, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Genève 4, Switzerland; (Y.N.); (L.W.); (F.K.); (S.N.)
| | - Corinne Loeuillet
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France; (C.C.); (C.L.); (C.A.); (P.F.R.)
| | - Lydia Wehrli
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Genève 4, Switzerland; (Y.N.); (L.W.); (F.K.); (S.N.)
| | - Françoise Kühne
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Genève 4, Switzerland; (Y.N.); (L.W.); (F.K.); (S.N.)
| | | | - Selima Fourati Ben Mustapha
- Centre d’Aide Médicale à la Procréation, Polyclinique les Jasmins, Centre Urbain Nord, Tunis 1003, Tunisia; (S.F.B.M.); (A.B.); (R.Z.)
| | - Amin Bouker
- Centre d’Aide Médicale à la Procréation, Polyclinique les Jasmins, Centre Urbain Nord, Tunis 1003, Tunisia; (S.F.B.M.); (A.B.); (R.Z.)
| | - Raoudha Zouari
- Centre d’Aide Médicale à la Procréation, Polyclinique les Jasmins, Centre Urbain Nord, Tunis 1003, Tunisia; (S.F.B.M.); (A.B.); (R.Z.)
| | | | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Genève 4, Switzerland; (Y.N.); (L.W.); (F.K.); (S.N.)
| | - Christophe Arnoult
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France; (C.C.); (C.L.); (C.A.); (P.F.R.)
| | - Pierre F. Ray
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France; (C.C.); (C.L.); (C.A.); (P.F.R.)
- UM GI-DPI, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Zine-Eddine Kherraf
- Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France; (C.C.); (C.L.); (C.A.); (P.F.R.)
- UM GI-DPI, CHU Grenoble Alpes, 38000 Grenoble, France;
- Correspondence: ; Tel.: +33-(0)4-7676-8303
| |
Collapse
|
33
|
Li Y, Wang Y, Wen Y, Zhang T, Wang X, Jiang C, Zheng R, Zhou F, Chen D, Yang Y, Shen Y. Whole-exome sequencing of a cohort of infertile men reveals novel causative genes in teratozoospermia that are chiefly related to sperm head defects. Hum Reprod 2021; 37:152-177. [PMID: 34791246 DOI: 10.1093/humrep/deab229] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Can whole-exome sequencing (WES) and in vitro validation studies identify new causative genes associated with teratozoospermia, particularly for sperm head defect? SUMMARY ANSWER We investigated a core group of infertile patients, including 82 cases with unexplained abnormal sperm head and 67 individuals with multiple morphological abnormalities of the sperm flagella (MMAF), and revealed rare and novel deleterious gene variants correlated with morphological abnormalities of the sperm head or tail defects. WHAT IS KNOWN ALREADY Teratozoospermia is one of the most common factors causing male infertility. Owing to high phenotypic variability, currently known genetic causes of teratozoospermia can only explain a rather minor component for patients with anomalous sperm-head shapes, and the agents responsible for atypical sperm head shapes remain largely unknown. STUDY DESIGN, SIZE, DURATION We executed WES analysis of a Chinese cohort of patients (N = 149) with teratozoospermia to identify novel genetic causes particularly for defective sperm head. We also sought to reveal the influence of different abnormalities of sperm morphology on ICSI outcome. PARTICIPANTS/MATERIALS, SETTING, METHODS In this study, a cohort of 149 infertile men (82 with abnormal sperm head and 67 with MMAF) were recruited. We implemented WES on infertile patients and analyzed the negative effects of the mutations of candidate genes on their protein conformations and/or expression. We also investigated the candidate genes' spatiotemporal expression/localization during spermatogenesis in both humans and mice, and explored their interactions with proteins that are known to be involved in sperm development. We also compared the ICSI outcomes of the affected individuals with various aberrations in sperm morphology. MAIN RESULTS AND THE ROLE OF CHANCE We identified rare and deleterious variants of piwi like RNA-mediated gene silencing 4 (PIWIL4: 1/82 patients, 1.21%), coiled-coil and C2 domain containing 1B (CC2D1B: 1/82 patients, 1.21%), cyclin B3 (CCNB3: 1/82 patients, 1.21%), KIAA1210 (KIAA1210: 2/82 patients, 2.43%) and choline phosphotransferase 1 (CHPT1: 1/82 patients, 1.21%), which are novel correlates of morphological abnormalities of the sperm head; functional evidence supports roles for all of these genes in sperm head formation. The mutations of septin 12 (SEPTIN12: 2/82 patients, 2.43%) are suggested to be associated with acrosome defects. We additionally observed novel causative mutations of dynein axonemal heavy chain 2 (DNAH2: 1/67 patients, 1.49%), dynein axonemal heavy chain 10 (DNAH10: 1/67 patients, 1.49%) and dynein axonemal heavy chain 12 (DNAH12: 1/67 patients, 1.49%) in patients with MMAF, and revealed a significantly lower fertilization rate of the abnormal sperm-head group compared to the MMAF group following ICSI. Consequently, our study also suggests that the mutations of PIWIL4 and CC2D1B might be circumvented by ICSI to a degree, and that CHPT1 and KIAA1210 loss-of-function variants might be associated with failed ICSI treatment. LIMITATIONS, REASONS FOR CAUTION In this study, we discovered the relationship between the genotype and phenotype of the novel causative genes of sperm head deformities in humans. However, the molecular mechanism of the relevant genes involved in sperm head development needs to be further illuminated in future research. Furthermore, evidence should be provided using knockout/knock-in mouse models for additional confirmation of the roles of these novel genes in spermatogenesis. WIDER IMPLICATIONS OF THE FINDINGS This cohort study of 149 Chinese infertile men documents novel genetic factors involved in teratozoospermia, particularly in anomalous sperm head formation. For the first time, we suggest that SEPTIN12 is related to human acrosomal hypoplasia, and that CCNB3 is a novel causative gene for globozoospermia in humans. We also uncovered variants in two genes-KIAA1210 and CHPT1associated with acrosomal biogenesis in patients with small or absent acrosomes. Additionally, it is postulated that loss-of-function mutations of PIWIL4 and CC2D1B have a contribution to the abnormal sperm-head formation. Furthermore, we are first to demonstrate the influence of different sperm morphologies on ICSI outcomes and indicates that the abnormal sperm head may play a significant role in fertilization failure. Our findings therefore provide valuable information for the diagnosis of teratozoospermia, particularly with respect to abnormalities of the sperm head. This will allow clinicians to adopt the optimal treatment strategy and to develop personalized medicine directly targeting these effects. STUDY FUNDING/COMPETING INTEREST(S) This work was financed by the West China Second University Hospital of Sichuan University (KS369 and KL042). The authors declare that they do not have any conflicts of interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yaqian Li
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Wang
- 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, China
| | - Yuting Wen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tao Zhang
- 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, China
| | - Xiaodong Wang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chuan Jiang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Zheng
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fan Zhou
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Daijuan Chen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 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, China
| | - Ying Shen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Castaneda JM, Shimada K, Satouh Y, Yu Z, Devlin DJ, Ikawa M, Matzuk MM. FAM209 associates with DPY19L2, and is required for sperm acrosome biogenesis and fertility in mice. J Cell Sci 2021; 134:272021. [PMID: 34471926 PMCID: PMC8627553 DOI: 10.1242/jcs.259206] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/31/2023] Open
Abstract
Infertility afflicts up to 15% of couples globally each year with men a contributing factor in 50% of these cases. Globozoospermia is a rare condition found in infertile men, which is characterized by defective acrosome biogenesis leading to the production of round-headed sperm. Here, we report that family with sequence similarity 209 (Fam209) is required for acrosome biogenesis in mouse sperm. FAM209 is a small transmembrane protein conserved among mammals. Loss of Fam209 results in fertility defects that are secondary to abnormalities in acrosome biogenesis during spermiogenesis, reminiscent of globozoospermia. Analysis of the FAM209 proteome identified DPY19L2, whose human orthologue is involved in the majority of globozoospermia cases. Although mutations in human and mouse Dpy19l2 have been shown to cause globozoospermia, no in vivo interacting partners of DPY19L2 have been identified until now. FAM209 colocalizes with DPY19L2 at the inner nuclear membrane to maintain the developing acrosome. Here, we identified FAM209 as the first interacting partner of DPY19L2, and the second protein that is essential for acrosome biogenesis that localizes to the inner nuclear membrane.
Collapse
Affiliation(s)
- Julio M Castaneda
- Research Institute for Microbial Diseases, Department of Experimental Genome Research, Osaka University, Osaka 5620031, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Department of Experimental Genome Research, Osaka University, Osaka 5620031, Japan
| | - Yuhkoh Satouh
- Institute for Molecular and Cellular Regulation, Department of Molecular and Cellular Biology, Gunma University, Gunma 3718512, Japan
| | - Zhifeng Yu
- Department of Pathology & Immunology and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Darius J Devlin
- Department of Pathology & Immunology and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Department of Experimental Genome Research, Osaka University, Osaka 5620031, Japan
| | - Martin M Matzuk
- Department of Pathology & Immunology and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
35
|
Morohoshi A, Miyata H, Oyama Y, Oura S, Noda T, Ikawa M. FAM71F1 binds to RAB2A and RAB2B and is essential for acrosome formation and male fertility in mice. Development 2021; 148:dev199644. [PMID: 34714330 PMCID: PMC8602946 DOI: 10.1242/dev.199644] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022]
Abstract
The acrosome is a cap-shaped, Golgi-derived membranous organelle that is located over the anterior of the sperm nucleus and highly conserved throughout evolution. Although morphological changes during acrosome biogenesis in spermatogenesis have been well described, the molecular mechanism underlying this process is still largely unknown. Family with sequence similarity 71, member F1 and F2 (FAM71F1 and FAM71F2) are testis-enriched proteins that contain a RAB2B-binding domain, a small GTPase involved in vesicle transport and membrane trafficking. Here, by generating mutant mice for each gene, we found that Fam71f1 is essential for male fertility. In Fam71f1-mutant mice, the acrosome was abnormally expanded at the round spermatid stage, likely because of enhanced vesicle trafficking. Mass spectrometry analysis after immunoprecipitation indicated that, in testes, FAM71F1 binds not only RAB2B, but also RAB2A. Further study suggested that FAM71F1 binds to the GTP-bound active form of RAB2A/B, but not the inactive form. These results indicate that a complex of FAM71F1 and active RAB2A/B suppresses excessive vesicle trafficking during acrosome formation.
Collapse
Affiliation(s)
- Akane Morohoshi
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yuki Oyama
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Seiya Oura
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Taichi Noda
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Division of Reproductive Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto 860-8555, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- Laboratory of Reproductive Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
36
|
Faja F, Pallotti F, Cargnelutti F, Senofonte G, Carlini T, Lenzi A, Lombardo F, Paoli D. Molecular Analysis of DPY19L2, PICK1 and SPATA16 in Italian Unrelated Globozoospermic Men. Life (Basel) 2021; 11:life11070641. [PMID: 34209343 PMCID: PMC8307282 DOI: 10.3390/life11070641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
This study aims to evaluate genetic contribution and sperm DNA fragmentation (SDF) in a cohort of 18 unrelated globozoospermic Italian men (Group G). Semen samples were assessed according to the WHO 2010 Laboratory Manual and compared with 31 fertile controls. We focused our genetic analysis on the exons of the main globozoospermia-associated genes, performing qualitative PCR to assess deletion of DPY19L2 and sequencing to detect mutations of SPATA16 and PICK1. SDF was evaluated using the TUNEL assay. In Group G, 10 patients had a complete form of globozoospermia, whereas 8 patients had a partial form. Molecular analysis revealed deletion of DPY19L2 in six of the patients, all of them with complete globozoospermia, while no mutations were found in the examined exons of PICK1 and SPATA16. TUNEL analysis showed a higher SDF% in Group G. Our findings confirm DPY19L2 defects as the most frequent genetic alteration in Italian patients contributing to globozoospermic phenotypes. Furthermore, spermatozoa with acrosomal defects could also display high levels of SDF as a possible consequence of abnormally remodeled chromatin. The possible effect on offspring of chromatin structure abnormalities and altered DNA integrity should be carefully evaluated by clinicians, especially regarding the feasibility and safety of artificial reproductive techniques, which represent the only treatment that allows these patients to conceive.
Collapse
|
37
|
Chen P, Saiyin H, Shi R, Liu B, Han X, Gao Y, Ye X, Zhang X, Sun Y. Loss of SPACA1 function causes autosomal recessive globozoospermia by damaging the acrosome-acroplaxome complex. Hum Reprod 2021; 36:2587-2596. [PMID: 34172998 DOI: 10.1093/humrep/deab144] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/08/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Is the sperm acrosome membrane-associated protein 1 (SPACA1) gene critical to human globozoospermia? SUMMARY ANSWER The biallelic loss-of-function (variant of SPACA1) causes globozoospermia as a result of acrosome-acroplaxome complex damage. WHAT IS KNOWN ALREADY SPACA1 expression decreases in patients with globozoospermia. Spaca1 gene-disrupted mice have abnormally shaped sperm heads that resemble those of human globozoospermia. STUDY DESIGN, SIZE, DURATION We recruited a consanguineous family with two brothers affected by infertility as a consequence of globozoospermia. The semen analysis data and ART outcomes were collected. Exome sequencing (ES) was used to identify potential pathogenic variants. Protein-protein interaction (PPI) technologies and proteomic analysis were utilized to explore the pathogenic mechanism. PARTICIPANTS/MATERIALS, SETTING, METHODS Two globozoospermic brothers and their consanguineous parents were recruited to identify the potential pathogenic variant through ES. A homozygous nonsense variant in the SPACA1 gene in both brothers inherited from the heterozygous parents was identified. Twenty normal fertile males were recruited as controls. Sperm ultrastructure was observed with transmission electron microscopy. Western blotting was performed to measure SPACA1 expression level in the sperm from the patients. Mass spectrometry (MS) analyses were used to identify differentially expressed proteins and to investigate proteins that interact with SPACA1. Co-immunoprecipitation (co-IP), yeast two-hybrid (Y2H) and immunofluorescence colocalization assays were used to confirm the PPI. MAIN RESULTS AND THE ROLE OF CHANCE A nonsense variant (NM_030960.2: c.53G>A; p. Trp18*) in the SPACA1 gene was identified as the pathogenic variant in a family with globozoospermia. Patient IV:1 and Patient IV:2 had a phenotype very similar to that of Spaca1 gene-disrupted mice. The nonsense variant in SPACA1 led to premature transcriptional termination in the signal peptide, which was confirmed by western blotting. MS-based proteomics analysis showed that eight interactors of SPACA1 were differentially expressed in the patients' sperm, including actin-like Protein 7A (ACTL7A), an important component of the acrosome-acroplaxome complex. The PPI of SPACA1 and ACTL7A was confirmed via co-IP and Y2H assays. Immunofluorescence showed that SPACA1 and ACTL7A colocalized in mature sperm, revealing that these proteins were coexpressed spatially. LIMITATIONS, REASONS FOR CAUTION Given the rarity of globozoospermia, only two patients from one family harbouring the SPACA1 variant were found. Future studies should evaluate SPACA1 variants in larger cohorts to corroborate this finding. WIDER IMPLICATIONS OF THE FINDINGS This study revealed that the SPACA1 gene was critical for globozoospermia, which expanded the spectrum of causative genes for globozoospermia. This study also provided evidence for ICSI clinical outcomes for patients with SPACA1-deficient globozoospermia, which may guide clinical treatment strategies. Furthermore, this study explored the pathogenesis of globozoospermia caused by SPACA1 deficiency. STUDY FUNDING/COMPETING INTEREST(S) This work was funded by the Precision Medical Research of National Key Research and Development Program (2018YFC1002400), National Natural Science Foundation of China (81873724), and Natural Science Foundation of Shanghai (20ZR1472700). The authors have no conflicts of interest to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Pingping Chen
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Paediatric Endocrinology/Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hexige Saiyin
- The State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Ruona Shi
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bin Liu
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Han
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuping Gao
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiantao Ye
- Department of Paediatric Endocrinology/Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Molecular Genetics Group, Shanghai Institute for Paediatric Research, Shanghai, China
| | - Xiaofei Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Metabolomics and Proteomics Center, Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yu Sun
- Department of Paediatric Endocrinology/Genetics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Molecular Genetics Group, Shanghai Institute for Paediatric Research, Shanghai, China
| |
Collapse
|
38
|
Azhar M, Altaf S, Uddin I, Cheng J, Wu L, Tong X, Qin W, Bao J. Towards Post-Meiotic Sperm Production: Genetic Insight into Human Infertility from Mouse Models. Int J Biol Sci 2021; 17:2487-2503. [PMID: 34326689 PMCID: PMC8315030 DOI: 10.7150/ijbs.60384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Declined quality and quantity of sperm is currently the major cause of patients suffering from infertility. Male germ cell development is spatiotemporally regulated throughout the whole developmental process. While it has been known that exogenous factors, such as environmental exposure, diet and lifestyle, et al, play causative roles in male infertility, recent progress has revealed abundant genetic mutations tightly associated with defective male germline development. In mammals, male germ cells undergo dramatic morphological change (i.e., nuclear condensation) and chromatin remodeling during post-meiotic haploid germline development, a process termed spermiogenesis; However, the molecular machinery players and functional mechanisms have yet to be identified. To date, accumulated evidence suggests that disruption in any step of haploid germline development is likely manifested as fertility issues with low sperm count, poor sperm motility, aberrant sperm morphology or combined. With the continually declined cost of next-generation sequencing and recent progress of CRISPR/Cas9 technology, growing studies have revealed a vast number of disease-causing genetic variants associated with spermiogenic defects in both mice and humans, along with mechanistic insights partially attained and validated through genetically engineered mouse models (GEMMs). In this review, we mainly summarize genes that are functional at post-meiotic stage. Identification and characterization of deleterious genetic variants should aid in our understanding of germline development, and thereby further improve the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Saba Altaf
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Islam Uddin
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Jinbao Cheng
- The 901th hospital of Joint logistics support Force of PLA, Anhui, China
| | - Limin Wu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Xianhong Tong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| |
Collapse
|
39
|
Xiong W, Shen C, Wang Z. The molecular mechanisms underlying acrosome biogenesis elucidated by gene-manipulated mice. Biol Reprod 2021; 105:789-807. [PMID: 34131698 DOI: 10.1093/biolre/ioab117] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 02/05/2023] Open
Abstract
Sexual reproduction requires the fusion of two gametes in a multistep and multifactorial process termed fertilization. One of the main steps that ensures successful fertilization is acrosome reaction. The acrosome, a special kind of organelle with a cap-like structure that covers the anterior portion of sperm head, plays a key role in the process. Acrosome biogenesis begins with the initial stage of spermatid development, and it is typically divided into four successive phases: the Golgi phase, cap phase, acrosome phase, and maturation phase. The run smoothly of above processes needs an active and specific coordination between the all kinds of organelles (endoplasmic reticulum, trans-golgi network and nucleus) and cytoplasmic structures (acroplaxome and manchette). During the past two decades, an increasingly genes have been discovered to be involved in modulating acrosome formation. Most of these proteins interact with each other and show a complicated molecular regulatory mechanism to facilitate the occurrence of this event. This Review focuses on the progresses of studying acrosome biogenesis using gene-manipulated mice and highlights an emerging molecular basis of mammalian acrosome formation.
Collapse
Affiliation(s)
- Wenfeng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
40
|
Manfrevola F, Guillou F, Fasano S, Pierantoni R, Chianese R. LINCking the Nuclear Envelope to Sperm Architecture. Genes (Basel) 2021; 12:genes12050658. [PMID: 33925685 PMCID: PMC8145172 DOI: 10.3390/genes12050658] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/24/2021] [Indexed: 12/11/2022] Open
Abstract
Nuclear architecture undergoes an extensive remodeling during spermatogenesis, especially at levels of spermatocytes (SPC) and spermatids (SPT). Interestingly, typical events of spermiogenesis, such as nuclear elongation, acrosome biogenesis, and flagellum formation, need a functional cooperation between proteins of the nuclear envelope and acroplaxome/manchette structures. In addition, nuclear envelope plays a key role in chromosome distribution. In this scenario, special attention has been focused on the LINC (linker of nucleoskeleton and cytoskeleton) complex, a nuclear envelope-bridge structure involved in the connection of the nucleoskeleton to the cytoskeleton, governing mechanotransduction. It includes two integral proteins: KASH- and SUN-domain proteins, on the outer (ONM) and inner (INM) nuclear membrane, respectively. The LINC complex is involved in several functions fundamental to the correct development of sperm cells such as head formation and head to tail connection, and, therefore, it seems to be important in determining male fertility. This review provides a global overview of the main LINC complex components, with a special attention to their subcellular localization in sperm cells, their roles in the regulation of sperm morphological maturation, and, lastly, LINC complex alterations associated to male infertility.
Collapse
Affiliation(s)
- Francesco Manfrevola
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania L. Vanvitelli, Via Costantinopoli 16, 80138 Napoli, Italy; (F.M.); (S.F.); (R.P.)
| | - Florian Guillou
- PRC, CNRS, IFCE, INRAE, University of Tours, 37380 Nouzilly, France;
| | - Silvia Fasano
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania L. Vanvitelli, Via Costantinopoli 16, 80138 Napoli, Italy; (F.M.); (S.F.); (R.P.)
| | - Riccardo Pierantoni
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania L. Vanvitelli, Via Costantinopoli 16, 80138 Napoli, Italy; (F.M.); (S.F.); (R.P.)
| | - Rosanna Chianese
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania L. Vanvitelli, Via Costantinopoli 16, 80138 Napoli, Italy; (F.M.); (S.F.); (R.P.)
- Correspondence:
| |
Collapse
|
41
|
Single-center thorough evaluation and targeted treatment of globozoospermic men. J Assist Reprod Genet 2021; 38:2073-2086. [PMID: 33877510 PMCID: PMC8417186 DOI: 10.1007/s10815-021-02191-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/07/2021] [Indexed: 11/10/2022] Open
Abstract
Purpose To characterize, by specific biomarkers and nucleic acid sequencing, the structural and genomic sperm characteristics of partial (PG) and complete globozoospermic (CG) men in order to identify the best reproductive treatment. Methods We assessed spermatozoa from 14 consenting men ultrastructurally, as well as for histone content, sperm chromatin integrity, and sperm aneuploidy. Additional genomic, transcriptomic, and proteomic evaluations were carried out to further characterize the CG cohort. The presence of oocyte-activating sperm cytosolic factor (OASCF) was measured by a phospholipase C zeta (PLCζ) immunofluorescence assay. Couples were treated in subsequent cycles either by conventional ICSI or by ICSI with assisted gamete treatment (AGT) using calcium ionophore (Ionomycin, 19657, Sigma-Aldrich, Saint Louis, MO, USA). Results Ultrastructural assessment confirmed complete acrosome deficiency in all spermatozoa from CG men. Histone content, sperm chromatin integrity, and sperm aneuploidy did not differ significantly between the PG (n = 4) and CG (n = 10) cohorts. PLCζ assessment indicated a positive presence of OASCF in 4 PG couples, who underwent subsequent ICSI cycles that yielded a 36.1% (43/119) fertilization with a 50% (2/4) clinical pregnancy and delivery rate. PLCζ assessment failed to detect OASCF for 8 CG patients who underwent 9 subsequent ICSI cycles with AGT, yielding a remarkable improvement of fertilization (39/97; 40.2%) (P = 0.00001). Embryo implantation (6/21; 28.6%) and clinical pregnancies (5/7; 71.4%) were also enhanced, resulting in 4 deliveries. Gene mutations (DPY19L2, SPATA16, PICK1) were identified in spermatozoa from CG patients. Additionally, CG patients unable to sustain a term pregnancy had gene mutations involved in zygote development (NLRP5) and postnatal development (BSX). CG patients who successfully sustained a pregnancy had a mutation (PIWIL1) related to sperm phenotype. PLCZ1 was both mutated and underexpressed in these CG patients, regardless of reproductive outcome. Conclusions Sperm bioassays and genomic studies can be used to characterize this gamete’s capacity to support embryonic development and to tailor treatments maximizing reproductive outcome. Supplementary Information The online version contains supplementary material available at 10.1007/s10815-021-02191-4.
Collapse
|
42
|
Choi H, Wang Z, Dean J. Sperm acrosome overgrowth and infertility in mice lacking chromosome 18 pachytene piRNA. PLoS Genet 2021; 17:e1009485. [PMID: 33831001 PMCID: PMC8057611 DOI: 10.1371/journal.pgen.1009485] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/20/2021] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
piRNAs are small non-coding RNAs required to maintain genome integrity and preserve RNA homeostasis during male gametogenesis. In murine adult testes, the highest levels of piRNAs are present in the pachytene stage of meiosis, but their mode of action and function remain incompletely understood. We previously reported that BTBD18 binds to 50 pachytene piRNA-producing loci. Here we show that spermatozoa in gene-edited mice lacking a BTBD18 targeted pachytene piRNA cluster on Chr18 have severe sperm head dysmorphology, poor motility, impaired acrosome exocytosis, zona pellucida penetration and are sterile. The mutant phenotype arises from aberrant formation of proacrosomal vesicles, distortion of the trans-Golgi network, and up-regulation of GOLGA2 transcripts and protein associated with acrosome dysgenesis. Collectively, our findings reveal central role of pachytene piRNAs in controlling spermiogenesis and male fertility.
Collapse
Affiliation(s)
- Heejin Choi
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| | - Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
43
|
Zakrzewski P, Lenartowska M, Buss F. Diverse functions of myosin VI in spermiogenesis. Histochem Cell Biol 2021; 155:323-340. [PMID: 33386429 PMCID: PMC8021524 DOI: 10.1007/s00418-020-01954-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Spermiogenesis is the final stage of spermatogenesis, a differentiation process during which unpolarized spermatids undergo excessive remodeling that results in the formation of sperm. The actin cytoskeleton and associated actin-binding proteins play crucial roles during this process regulating organelle or vesicle delivery/segregation and forming unique testicular structures involved in spermatid remodeling. In addition, several myosin motor proteins including MYO6 generate force and movement during sperm differentiation. MYO6 is highly unusual as it moves towards the minus end of actin filaments in the opposite direction to other myosin motors. This specialized feature of MYO6 may explain the many proposed functions of this myosin in a wide array of cellular processes in animal cells, including endocytosis, secretion, stabilization of the Golgi complex, and regulation of actin dynamics. These diverse roles of MYO6 are mediated by a range of specialized cargo-adaptor proteins that link this myosin to distinct cellular compartments and processes. During sperm development in a number of different organisms, MYO6 carries out pivotal functions. In Drosophila, the MYO6 ortholog regulates actin reorganization during spermatid individualization and male KO flies are sterile. In C. elegans, the MYO6 ortholog mediates asymmetric segregation of cytosolic material and spermatid budding through cytokinesis, whereas in mice, this myosin regulates assembly of highly specialized actin-rich structures and formation of membrane compartments to allow the formation of fully differentiated sperm. In this review, we will present an overview and compare the diverse function of MYO6 in the specialized adaptations of spermiogenesis in flies, worms, and mammals.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
44
|
Beurois J, Cazin C, Kherraf ZE, Martinez G, Celse T, Touré A, Arnoult C, Ray PF, Coutton C. Genetics of teratozoospermia: Back to the head. Best Pract Res Clin Endocrinol Metab 2020; 34:101473. [PMID: 33183966 DOI: 10.1016/j.beem.2020.101473] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Spermatozoa are polarized cells with a head and a flagellum joined by the connecting piece. Head integrity is critical for normal sperm function, and head defects consistently lead to male infertility. Abnormalities of the sperm head are among the most severe and characteristic sperm defects. Patients presenting with a monomorphic head sperm defects such as globozoospermia or marcrozoospermia were analyzed permitting to identify several key genes for spermatogenesis such as AURKC and DPY19L2. The study of patients with other specific sperm head defects such as acephalic spermatozoa have also enabled the identification of new infertility genes such as SUN5. Here, we review the genetic causes leading to morphological defects of sperm head. Advances in the genetics of male infertility are necessary to improve the management of infertility and will pave the road towards future strategies of treatments, especially for patients with the most severe phenotype as sperm head defects.
Collapse
Affiliation(s)
- Julie Beurois
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France
| | - Caroline Cazin
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France
| | - Zine-Eddine Kherraf
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France
| | - Guillaume Martinez
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France; CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, France
| | - Tristan Celse
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France; CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, France
| | - Aminata Touré
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France
| | - Christophe Arnoult
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France
| | - Pierre F Ray
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France
| | - Charles Coutton
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, 38000, Grenoble, France; CHU de Grenoble, UM GI-DPI, Grenoble, F-38000, France; CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, France.
| |
Collapse
|
45
|
Kmonickova V, Frolikova M, Steger K, Komrskova K. The Role of the LINC Complex in Sperm Development and Function. Int J Mol Sci 2020; 21:E9058. [PMID: 33260574 PMCID: PMC7730847 DOI: 10.3390/ijms21239058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 11/23/2022] Open
Abstract
The LINC (LInker of Nucleoskeleton and Cytoskeleton) complex is localized within the nuclear envelope and consists of SUN (Sad1/UNc84 homology domain-containing) proteins located in the inner nuclear membrane and KASH (Klarsicht/Anc1/Syne1 homology domain-containing) proteins located in the outer nuclear membrane, hence linking nuclear with cytoplasmic structures. While the nucleoplasm-facing side acts as a key player for correct pairing of homolog chromosomes and rapid chromosome movements during meiosis, the cytoplasm-facing side plays a pivotal role for sperm head development and proper acrosome formation during spermiogenesis. A further complex present in spermatozoa is involved in head-to-tail coupling. An intact LINC complex is crucial for the production of fertile sperm, as mutations in genes encoding for complex proteins are known to be associated with male subfertility in both mice and men. The present review provides a comprehensive overview on our current knowledge of LINC complex subtypes present in germ cells and its central role for male reproduction. Future studies on distinct LINC complex components are an absolute requirement to improve the diagnosis of idiopathic male factor infertility and the outcome of assisted reproduction.
Collapse
Affiliation(s)
- Vera Kmonickova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic; (V.K.); (M.F.)
| | - Michaela Frolikova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic; (V.K.); (M.F.)
| | - Klaus Steger
- Department of Urology, Pediatric Urology and Andrology, Molecular Andrology, Justus-Liebig University, 35392 Giessen, Germany;
| | - Katerina Komrskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic; (V.K.); (M.F.)
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, 128 44 Prague 2, Czech Republic
| |
Collapse
|
46
|
She ZY, Yu KW, Wei YL, Zhong N, Lin Y. Kinesin-7 CENP-E regulates the formation and structural maintenance of the acrosome. Cell Tissue Res 2020; 383:1167-1182. [PMID: 33237480 DOI: 10.1007/s00441-020-03341-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 11/05/2020] [Indexed: 12/23/2022]
Abstract
The acrosome is a special organelle that develops from the Golgi apparatus and the endolysosomal compartment in the spermatids. Centromere protein E (CENP-E) is an essential kinesin motor in chromosome congression and alignment. This study is aimed at investigating the roles and mechanisms of kinesin-7 CENP-E in the formation of the acrosome during spermatogenesis. Male ICR mice are injected with GSK923295 for long-term inhibition of CENP-E. Chemical inhibition and siRNA-mediated knockdown of CENP-E are carried out in the GC-2 spd cells. The morphology of the acrosomes is determined by the HE staining, immunofluorescence, and transmission electron microscopy. We have identified CENP-E is a key factor in the formation and structural maintenance of the acrosome during acrosome biogenesis. Long-term inhibition of CENP-E by GSK923295 results in the asymmetric acrosome and the dispersed acrosome. CENP-E depletion leads to the malformation of the Golgi complex and abnormal targeting of the PICK1- and PIST-positive Golgi-associated vesicles. Our findings uncover an essential role of CENP-E in membrane trafficking and structural organization of the acrosome in the spermatids during spermatogenesis. Our results shed light on the molecular mechanisms involved in vesicle trafficking and architecture maintenance of the acrosome.
Collapse
Affiliation(s)
- Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China. .,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China.
| | - Kai-Wei Yu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350011, Fujian, China.,Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ning Zhong
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Yang Lin
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| |
Collapse
|
47
|
Vatin M, Girault MS, Firlej V, Marchiol C, Ialy-Radio C, Montagutelli X, Vaiman D, Barbaux S, Ziyyat A. Identification of a New QTL Region on Mouse Chromosome 1 Responsible for Male Hypofertility: Phenotype Characterization and Candidate Genes. Int J Mol Sci 2020; 21:ijms21228506. [PMID: 33198087 PMCID: PMC7697627 DOI: 10.3390/ijms21228506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.
Collapse
Affiliation(s)
- Magalie Vatin
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Marie-Sophie Girault
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Virginie Firlej
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Carmen Marchiol
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Côme Ialy-Radio
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | | | - Daniel Vaiman
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Sandrine Barbaux
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
| | - Ahmed Ziyyat
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014 Paris, France; (M.V.); (M.-S.G.); (V.F.); (C.M.); (C.I.-R.); (D.V.); (S.B.)
- Service d’histologie, d’embryologie, Biologie de la Reproduction, AP-HP, Hôpital Cochin, F-75014 Paris, France
- Correspondence:
| |
Collapse
|
48
|
Jiao SY, Yang YH, Chen SR. Molecular genetics of infertility: loss-of-function mutations in humans and corresponding knockout/mutated mice. Hum Reprod Update 2020; 27:154-189. [PMID: 33118031 DOI: 10.1093/humupd/dmaa034] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Infertility is a major issue in human reproductive health, affecting an estimated 15% of couples worldwide. Infertility can result from disorders of sex development (DSD) or from reproductive endocrine disorders (REDs) with onset in infancy, early childhood or adolescence. Male infertility, accounting for roughly half of all infertility cases, generally manifests as decreased sperm count (azoospermia or oligozoospermia), attenuated sperm motility (asthenozoospermia) or a higher proportion of morphologically abnormal sperm (teratozoospermia). Female infertility can be divided into several classical types, including, but not limited to, oocyte maturation arrest, premature ovarian insufficiency (POI), fertilization failure and early embryonic arrest. An estimated one half of infertility cases have a genetic component; however, most genetic causes of human infertility are currently uncharacterized. The advent of high-throughput sequencing technologies has greatly facilitated the identification of infertility-associated gene mutations in patients over the past 20 years. OBJECTIVE AND RATIONALE This review aims to conduct a narrative review of the genetic causes of human infertility. Loss-of-function mutation discoveries related to human infertility are summarized and further illustrated in tables. Corresponding knockout/mutated animal models of causative genes for infertility are also introduced. SEARCH METHODS A search of the PubMed database was performed to identify relevant studies published in English. The term 'mutation' was combined with a range of search terms related to the core focus of the review: infertility, DSD, REDs, azoospermia or oligozoospermia, asthenozoospermia, multiple morphological abnormalities of the sperm flagella (MMAF), primary ciliary dyskinesia (PCD), acephalic spermatozoa syndrome (ASS), globozoospermia, teratozoospermia, acrosome, oocyte maturation arrest, POI, zona pellucida, fertilization defects and early embryonic arrest. OUTCOMES Our search generated ∼2000 records. Overall, 350 articles were included in the final review. For genetic investigation of human infertility, the traditional candidate gene approach is proceeding slowly, whereas high-throughput sequencing technologies in larger cohorts of individuals is identifying an increasing number of causative genes linked to human infertility. This review provides a wide panel of gene mutations in several typical forms of human infertility, including DSD, REDs, male infertility (oligozoospermia, MMAF, PCD, ASS and globozoospermia) and female infertility (oocyte maturation arrest, POI, fertilization failure and early embryonic arrest). The causative genes, their identified mutations, mutation rate, studied population and their corresponding knockout/mutated mice of non-obstructive azoospermia, MMAF, ASS, globozoospermia, oocyte maturation arrest, POI, fertilization failure and early embryonic arrest are further illustrated by tables. In this review, we suggest that (i) our current knowledge of infertility is largely obtained from knockout mouse models; (ii) larger cohorts of clinical cases with distinct clinical characteristics need to be recruited in future studies; (iii) the whole picture of genetic causes of human infertility relies on both the identification of more mutations for distinct types of infertility and the integration of known mutation information; (iv) knockout/mutated animal models are needed to show whether the phenotypes of genetically altered animals are consistent with findings in human infertile patients carrying a deleterious mutation of the homologous gene; and (v) the molecular mechanisms underlying human infertility caused by pathogenic mutations are largely unclear in most current studies. WILDER IMPLICATIONS It is important to use our current understanding to identify avenues and priorities for future research in the field of genetic causes of infertility as well as to apply mutation knowledge to risk prediction, genetic diagnosis and potential treatment for human infertility.
Collapse
Affiliation(s)
- Shi-Ya Jiao
- Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Yi-Hong 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, 610041 Chengdu, China
| | - Su-Ren Chen
- Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| |
Collapse
|
49
|
Celse T, Cazin C, Mietton F, Martinez G, Martinez D, Thierry-Mieg N, Septier A, Guillemain C, Beurois J, Clergeau A, Mustapha SFB, Kharouf M, Zoghmar A, Chargui A, Papaxanthos A, Dorphin B, Foliguet B, Triki C, Sifer C, Lauton D, Tachdjian G, Schuler G, Lejeune H, Puechberty J, Bessonnat J, Pasquier L, Mery L, Poulain M, Chaabouni M, Sermondade N, Cabry R, Benbouhadja S, Veau S, Frapsauce C, Mitchell V, Achard V, Satre V, Hennebicq S, Zouari R, Arnoult C, Kherraf ZE, Coutton C, Ray PF. Genetic analyses of a large cohort of infertile patients with globozoospermia, DPY19L2 still the main actor, GGN confirmed as a guest player. Hum Genet 2020; 140:43-57. [PMID: 33108537 DOI: 10.1007/s00439-020-02229-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/15/2020] [Indexed: 12/28/2022]
Abstract
Globozoospermia is a rare phenotype of primary male infertility inducing the production of round-headed spermatozoa without acrosome. Anomalies of DPY19L2 account for 50-70% of all cases and the entire deletion of the gene is by far the most frequent defect identified. Here, we present a large cohort of 69 patients with 20-100% of globozoospermia. Genetic analyses including multiplex ligation-dependent probe amplification, Sanger sequencing and whole-exome sequencing identified 25 subjects with a homozygous DPY19L2 deletion (36%) and 14 carrying other DPY19L2 defects (20%). Overall, 11 deleterious single-nucleotide variants were identified including eight novel and three already published mutations. Patients with a higher rate of round-headed spermatozoa were more often diagnosed and had a higher proportion of loss of function anomalies, highlighting a good genotype phenotype correlation. No gene defects were identified in patients carrying < 50% of globozoospermia while diagnosis efficiency rose to 77% for patients with > 50% of globozoospermia. In addition, results from whole-exome sequencing were scrutinized for 23 patients with a DPY19L2 negative diagnosis, searching for deleterious variants in the nine other genes described to be associated with globozoospermia in human (C2CD6, C7orf61, CCDC62, CCIN, DNAH17, GGN, PICK1, SPATA16, and ZPBP1). Only one homozygous novel truncating variant was identified in the GGN gene in one patient, confirming the association of GGN with globozoospermia. In view of these results, we propose a novel diagnostic strategy focusing on patients with at least 50% of globozoospermia and based on a classical qualitative PCR to detect DPY19L2 homozygous deletions. In the absence of the latter, we recommend to perform whole-exome sequencing to search for defects in DPY19L2 as well as in the other previously described candidate genes.
Collapse
Affiliation(s)
- Tristan Celse
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Caroline Cazin
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Flore Mietton
- CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Guillaume Martinez
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000, Grenoble, France
| | | | | | - Amandine Septier
- Université Grenoble Alpes, CNRS, TIMC-IMAG, 38000, Grenoble, France
| | - Catherine Guillemain
- Pôle Femmes-Parents-Enfants, Centre Clinico-Biologique AMP-CECOS, Plateforme Cancer et Fertilité ONCOPACA-Corse, Assistance-Publique des Hôpitaux de Marseille (AP-HM), Marseille, France.,Aix Marseille University, INSERM, MMG, UMR_S 1251, Marseille, France
| | - Julie Beurois
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France
| | | | | | - Mahmoud Kharouf
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain Nord, 1003, Tunis, Tunisia
| | - Abdelali Zoghmar
- Reproduction Sciences and Surgery Clinique, Ibn Rochd, Constantine, Algeria
| | - Ahmed Chargui
- Faculté de Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire (CHU) Cochin, Service d'Histologie-Embryologie-Biologie de la Reproduction, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Aline Papaxanthos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Bordeaux University Hospital, Bordeaux, France
| | | | - Bernard Foliguet
- Toxicology and Molecular Biology, Institute Jean Lamour UMR 7198 du CNRS, Université de Lorraine, 54000, Nancy, France
| | - Chema Triki
- Centre d'AMP, Clinique Hannibal, Les Berges du Lac, 1053, Tunis, Tunisia
| | - Christophe Sifer
- Service de Biologie de la Reproduction, d'Histo-Embryologie et Cytogénétique, Hôpital Jean-Verdier, Avenue du 14 Juillet, 93140, Bondy, France
| | - Dominique Lauton
- Department of Endocrinology, Diabetes, Nutrition, Montpellier University Hospital, Montpellier, France
| | - Gérard Tachdjian
- UMR 967, INSERM, Service d'Histologie Embryologie et Cytogénétique, Hôpitaux Universitaires Paris-Sud, AP-HP, Clamart, France
| | | | - Hervé Lejeune
- Reproductive Medicine Department, Hospices Civils de Lyon, Lyon, France
| | - Jacques Puechberty
- Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Hôpital Arnaud de Villeneuve, CHU de Montpellier, Université Montpelier, Montpellier, France
| | - Julien Bessonnat
- CHU de Grenoble, UF de Biologie de la Procréation, 38000, Grenoble, France
| | - Laurent Pasquier
- Service de Génétique Clinique, CLAD Ouest, CHU Rennes, Rennes, France
| | - Lionel Mery
- Service de Médecine de la Reproduction, CHU de Saint-Étienne, Hôpital Nord, 42055, Saint-Étienne Cedex 2, France
| | - Marine Poulain
- Department of Obstetrics and Gynecology, Hôpital Foch, Université de Paris Ouest (UVSQ), Suresnes, France
| | - Myriam Chaabouni
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain Nord, 1003, Tunis, Tunisia
| | - Nathalie Sermondade
- Service de Biologie de la Reproduction-CECOS, Hôpital Tenon, AP-HP, 75020, Paris, France
| | - Rosalie Cabry
- Department of Obstetrics, Gynaecology and Reproductive Medicine, Picardie University Jules Verne, Amiens University Medical Centre, Amiens, France
| | - Sebti Benbouhadja
- Reproduction Sciences and Surgery Clinique, Ibn Rochd, Constantine, Algeria
| | - Ségolène Veau
- CHU, Centre d'AMP-CECOS, University Rennes, 16 Boulevard de Bulgarie, 35000, Rennes, France
| | - Cynthia Frapsauce
- CHU Bretonneau, Médecine et Biologie de la Reproduction-CECOS, Tours, France
| | - Valérie Mitchell
- EA 4308, Department of Reproductive Biology and Spermiology-CECOS Lille, University Medical Center, 59037, Lille, France
| | - Vincent Achard
- CECOS-Laboratoire de Biologie de la Reproduction, Pôle de Gynécologie Obstétrique et Reproduction (Gynépôle), Assistance Publique-Hôpitaux de Marseille (AP-HM) la Conception, 13005, Marseille, France.,Centre Clinico-Biologique d'Assistance Médicale à la Procréation, Pôle de Gynécologie Obstétrique et Reproduction (Gynépôle), Assistance Publique-Hôpitaux de Marseille (AP-HM) la Conception, 13005, Marseille, France.,Faculté de Médecine, Institut Méditerranéen de Biodiversité et d'Écologie (IMBE UMR 7263), Equipe Biogénotoxicologie, Santé Humaine et Environnement, Aix Marseille Université, CNRS, IRD, Université Avignon, 27, Boulevard Jean-Moulin, 13385, Marseille Cedex 5, France
| | - Veronique Satre
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000, Grenoble, France
| | - Sylviane Hennebicq
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU de Grenoble, UF de Biologie de la Procréation, 38000, Grenoble, France
| | - Raoudha Zouari
- Pôle Femmes-Parents-Enfants, Centre Clinico-Biologique AMP-CECOS, Plateforme Cancer et Fertilité ONCOPACA-Corse, Assistance-Publique des Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Christophe Arnoult
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France
| | - Zine-Eddine Kherraf
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Charles Coutton
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France.,CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000, Grenoble, France
| | - Pierre F Ray
- Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, 38000, Grenoble, France. .,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France.
| |
Collapse
|
50
|
Han Y, Liang C, Yu Y, Manthari RK, Cheng C, Tan Y, Li X, Tian X, Fu W, Yang J, Yang W, Xing Y, Wang J, Zhang J. Chronic arsenic exposure lowered sperm motility via impairing ultra-microstructure and key proteins expressions of sperm acrosome and flagellum formation during spermiogenesis in male mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 734:139233. [PMID: 32460071 DOI: 10.1016/j.scitotenv.2020.139233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 06/11/2023]
Abstract
Arsenic (As) poisoning and its potential reproductive functional lesions are a global environmental concern. Recent studies shown that spermiogenesis tends to be a major target process in arsenic-induced male infertility, however, the underlying mechanisms are not fully illuminated. In the present study, 32 fertility related indices including sperm motility, dynamic acrosome formation and sperm flagellum during spermiogenesis in testes were evaluated in adult male mice treated with 0, 0.2, 2, and 20 ppm As2O3 via drinking water for 180 consecutive days. The results showed that out of 32 indices, 11, 25, and 29 indicators were changed statistically by 0.2-, 2-, and 20- ppm As2O3 treatment compared to the controls (0 ppm As2O3), respectively, which reveals a significant dose-dependent relationship. For details, sperm motilities were significantly decreased by 18.85%, 32.47% and 29.53% in three As2O3 treatment groups compared to the control group. Meanwhile, the ultra-structures of acrosome formation and sperm flagellum in testes have been altered by chronic arsenic exposure. Furthermore, arsenic decreased the mRNA expressions of 11 out of 13 genes associated with acrosome biosynthesis and 11 out of 12 genes related to flagellum formation in testes, particularly, down-regulated DPY19L2, AKAP3, AKAP4, CFAP44 and SPAG16 were further confirmed at the protein levels by western blotting. Taken together, chronic arsenic exposure declines male fertility by disorganizing dynamic acrosome and flagellum formation in testes. Especially, DPY19L2, AKAP3, AKAP4, CFAP44, and SPAG16 maybe the potential targets in this process. These results may offer not only a new insight to the mechanism of arsenic-induced male reproductive toxicity, but also provide a clue for the diagnosis and therapy of arseniasis.
Collapse
Affiliation(s)
- Yongli Han
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Chen Liang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yuxiang Yu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Ram Kumar Manthari
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Chenkai Cheng
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yanjia Tan
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Xiang Li
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Xiaolin Tian
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Weixiang Fu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jie Yang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Wei Yang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yin Xing
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jianhai Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China.
| |
Collapse
|