1
|
Huang M, Zhou J, Yihao Z, Luo K, Zheng S, Tang H, Wu Y, Xuan R, Huang Y, Li J, Xiong R, Fang X, Wang L, Gong Y, Miao J, Tan H, Wang Y, Wu L, Ouyang J, Yan X, Chen H. Whole genome sequencing revealed genetic structure, domestication, and selection of Chinese indigenous ducks. Int J Biol Macromol 2025; 306:141724. [PMID: 40043989 DOI: 10.1016/j.ijbiomac.2025.141724] [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: 10/07/2024] [Revised: 01/15/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
The genetic architecture and domestication history of Chinese indigenous ducks, particularly those with distinct traits like the Longsheng duck (LSD), are not well characterized. This study used whole-genome resequencing data from 540 ducks across 30 populations to explore the genetic diversity and structure of these ducks. Our findings suggest extensive interspecific hybridization between mallard and spot-billed duck. Comparing with other Chinese ducks, LSD is a distinct breed with moderate genetic diversity. Selective signal analysis identified several key genes impacting neural development, fat metabolism, immunity, and circadian rhythms like SLC25A20 and PER2. These genes showed strong selective pressures that parallelled other domestication processes. Additionally, EDNRB2 was identified as a potential gene influencing the unique coloration of LSD's plumage, bill, and webbed feet, associated with distinct mutations in non-coding regions. Comparative analysis with other indigenous breeds further pinpointed genes associated with LSD-specific traits, including plumage color, reproductive capabilities, and fat deposition, such as MITF, SPATA2, EIF2S2, PLIN3, ATP1B1, and CCDC80. Our findings clarify the population genetics of Chinese indigenous ducks. They also highlight key genes and mutations that shape the unique characteristics of LSD. These insights pave the way for further research into the genetic resources and domestication patterns of Chinese ducks.
Collapse
Affiliation(s)
- Min Huang
- College of Animal Sciences & Technology, Zhejiang A&F University, Hangzhou, China
| | - Jing Zhou
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Zhu Yihao
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Keyi Luo
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Sumei Zheng
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Hongbo Tang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yan Wu
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Rui Xuan
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yuxuan Huang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiawei Li
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Rui Xiong
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Xinyan Fang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Lei Wang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yujie Gong
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Junjie Miao
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Hongli Tan
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yanan Wang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Liping Wu
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jing Ouyang
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Xueming Yan
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| | - Hao Chen
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
2
|
Xie CT, Zhang HL, Li Y, Li Q, Wen YX, Liu JY, Han F. Single-cell RNA-seq and pathological phenotype reveal the functional atlas and precise roles of Sox30 in testicular cell development and differentiation. Cell Death Dis 2025; 16:110. [PMID: 39971903 PMCID: PMC11840104 DOI: 10.1038/s41419-025-07442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Sox30 has recently been demonstrated to be a key regulator of spermatogenesis. However, the precise roles of Sox30 in the testis remain largely unclear. Here, the specific functions of Sox30 in testicular cells were determined by single-cell sequencing and confirmed via pathological analyses. Sox30 loss appears to damage all testicular cells to different extents. Sox30 chiefly drives the differentiation of primary spermatocytes. Sox30 deficiency causes spermatocyte arrest at the early phase of meiosis I, with nearly no normally developing second spermatocytes and three new spermatocyte -subclusters emerging. In addition, Sox30 seems to play important roles in the mature phenotypes of Sertoli and Leydig cells, and the proliferation and differentiation of spermatogonia. The developmental trajectory of germ cells begins with spermatogonia and splits into two different spermatocyte branches, with Sox30-null spermatocytes and wild-type spermatocytes placed at divergent ends. An opposite developmental trajectory of spermatocyte subclusters is observed, followed by incomplete development of spermatid subclusters in Sox30-null mice. Sox30 deficiency clearly alters the intercellular cross-talk of major testicular cells and dysregulates the transcription factor networks primarily involved in cell proliferation and differentiation. Mechanistically, Sox30 appears to have similar terminal functions that are involved mainly in spermatogenic development and differentiation among major testicular cells, and Sox30 performs these similar crucial roles through preferential regulation of different signalling pathways. Our study describes the exact functions of Sox30 in testicular cell development and differentiation and highlights the primary roles of Sox30 in the early meiotic phase of germ cells.
Collapse
Affiliation(s)
- Cheng-Ting Xie
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China
| | - Hui-Lian Zhang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China
| | - Yi Li
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China
| | - Qian Li
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China
| | - Yi-Xian Wen
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China
| | - Jin-Yi Liu
- Institute of Toxicology, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| | - Fei Han
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education, Chongqing, 400016, China.
| |
Collapse
|
3
|
Cheung FKM, Feng CWA, Crisp C, Mishina Y, Spiller CM, Bowles J. BMP and STRA8 act collaboratively to ensure correct mitotic-to-meiotic transition in the fetal mouse ovary. Development 2025; 152:DEV204227. [PMID: 39817676 PMCID: PMC11829761 DOI: 10.1242/dev.204227] [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: 07/05/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
A successful mitosis-to-meiosis transition in germ cells is essential for fertility in sexually reproducing organisms. In mice and humans, it has been established that expression of STRA8 is crucial for meiotic onset in both sexes. Here, we show that BMP signalling is also essential, not for STRA8 induction but for correct meiotic progression in female mouse fetal germ cells. Largely in agreement with evidence from primordial germ cell-like cells (PGCLCs) in vitro, germ cell-specific deletion of BMP receptor 1A (BMPR1A; ALK3) caused aberrant retention of pluripotency marker OCT4 and meiotic progression was compromised; however, the timely onset of Stra8 and STRA8 expression was unaffected. Comparing the transcriptomes of Bmpr1a-cKO and Stra8-null models, we reveal interplay between the effects of BMP signalling and STRA8 function. Our results verify a role for BMP signalling in instructing germ cell meiosis in female mice in vivo, and shed light on the regulatory mechanisms underlying fetal germ cell development.
Collapse
Affiliation(s)
- Fiona K M Cheung
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chun-Wei Allen Feng
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Clare Crisp
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yuji Mishina
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cassy M Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
4
|
Li J, Liang J, Wang M, Jiang Y, Li W, Huang M, Huang Y, Xie Y, Chen J, Chen T. Full-length transcriptome analysis of male and female gonads in Japanese Eel (Anguilla japonica). BMC Genomics 2025; 26:89. [PMID: 39885385 PMCID: PMC11783869 DOI: 10.1186/s12864-025-11279-5] [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: 08/08/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND The Japanese eel (Anguilla japonica) holds significant economic value in East Asia, but limitations in understanding its reproductive biology have hindered advancements in artificial breeding techniques. Previous research has primarily focused on conserved sex differentiation genes, offering limited insights into the broader molecular mechanisms driving gonadal development and sexual dimorphism. To address these limitations, this study aims to investigate key genes and pathways involved in gonadal development through a comprehensive transcriptomic analysis of male and female eel gonads. RESULTS PacBio Iso-Seq and Illumina RNA-Seq technologies were combined to conduct a full-length transcriptome analysis of male and female Japanese eel gonads at a post-differentiation, pre-maturation stage. A total of 24,661 unigenes were identified in ovaries and 15,023 in testes, along with genomic regulatory elements such as transcription factors, simple sequence repeats, and long non-coding RNAs. Additionally, 1,210 differentially expressed genes were detected. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed significant pathways involved in cell cycle regulation, metabolic processes, apoptosis, and hormone activity. Notably, several reproductive-related genes, including bambi, ccnb1, cdc20, gdf9, prlh, ccdc39, chrebp, tspo, syce3, and ngb, demonstrated significant dimorphic expression in eel gonads. CONCLUSIONS This study provides valuable insights into the molecular mechanisms of gonadal differentiation and sexual dimorphism in Japanese eels. The findings expand the genetic resources available for the eel breeding industry and could facilitate the development of improved artificial breeding techniques focused on reproductive development.
Collapse
Affiliation(s)
- Jiangling Li
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Jingjie Liang
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Mengyang Wang
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Yuewen Jiang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wen Li
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Mingxi Huang
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Yan Huang
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Yangjie Xie
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Jianchun Chen
- Xiamen Institute of Marine and Fisheries, Xiamen, Fujian, 361013, China
| | - Tiansheng Chen
- State Key Laboratory of Mariculture Breeding; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education;Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College of Jimei University, Xiamen, 361021, China.
| |
Collapse
|
5
|
García-Martínez OI, Geisinger A, de Los Santos E, Santiñaque FF, Folle GA, Pórfido JL, Meikle MN, Schlapp G, Crispo M, Benavente R, Rodríguez-Casuriaga R. Mouse modeling of familial human SYCE1 c.197-2A>G splice site mutation leads to meiotic recombination failure and non-obstructive azoospermia. Mol Hum Reprod 2025; 31:gaaf002. [PMID: 39909494 DOI: 10.1093/molehr/gaaf002] [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: 06/18/2024] [Revised: 12/24/2024] [Indexed: 02/07/2025] Open
Abstract
Infertility affects a considerable number of couples at reproductive age, with an incidence of 10-15%. Approximately 25% of cases are classified as idiopathic infertility. Often, errors during the meiotic stage appear to be related to idiopathic infertility. A crucial component during the first meiotic prophase is the synaptonemal complex (SC), which plays a fundamental role in homologous chromosome pairing and meiotic recombination. In many studies with infertile patients, mutations affecting SC-coding genes have been identified. The generation of humanized models has high physiological relevance, helping to clarify the molecular bases of pathology, which in turn is essential for the development of therapeutic procedures. Here, we report the generation and characterization of genetically modified mice carrying a mutation equivalent to SYCE1 c.197-2A>G, previously found in male infertile patients, aiming to determine the actual effects of this mutation on reproductive capacity and to study the underlying molecular mechanisms. Homozygous mutants were infertile. SYCE1 protein was not detected and Syce1 transcript presented minimal levels, suggesting transcript degradation underlying the infertility mechanism. Additionally, homozygous mutants showed impaired homologous chromosome synapsis, meiotic arrest before the pachytene stage, and increased apoptosis of meiotic cells. This study validates the variant as pathogenic and causative of infertility, since the observed dramatic phenotype was attributable to this single homozygous point mutation, when compared to WT and heterozygous littermates. Moreover, although this homozygous point mutation has been only found in infertile men thus far, we anticipate that if it were present in women, it would cause infertility as well, as homozygous female mice also exhibited an infertility phenotype.
Collapse
Affiliation(s)
- Omar Ignacio García-Martínez
- Laboratory of Molecular Biology of Reproduction, Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Adriana Geisinger
- Laboratory of Molecular Biology of Reproduction, Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Biochemistry-Molecular Biology, Facultad de Ciencias, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Eliana de Los Santos
- Laboratory of Molecular Biology of Reproduction, Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | | | - Gustavo A Folle
- Flow Cytometry and Cell Sorting Core, IIBCE, Montevideo, Uruguay
| | - Jorge Luis Pórfido
- Laboratory Animal Biotechnology Unit (UBAL), Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - María Noel Meikle
- Laboratory Animal Biotechnology Unit (UBAL), Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Geraldine Schlapp
- Laboratory Animal Biotechnology Unit (UBAL), Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Martina Crispo
- Laboratory Animal Biotechnology Unit (UBAL), Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Ricardo Benavente
- Laboratory of Molecular Biology of Reproduction, Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Rosana Rodríguez-Casuriaga
- Laboratory of Molecular Biology of Reproduction, Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| |
Collapse
|
6
|
Kursel LE, Goktepe K, Rog O. The structural role of Skp1 in the synaptonemal complex is conserved in nematodes. Genetics 2024; 228:iyae153. [PMID: 39293001 PMCID: PMC11538402 DOI: 10.1093/genetics/iyae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
The synaptonemal complex (SC) is a meiotic interface that assembles between parental chromosomes and is essential for gamete formation. While the dimensions and ultrastructure of the SC are conserved across eukaryotes, its protein components are highly divergent. Recently, an unexpected component of the SC has been described in the nematode Caenorhabditis elegans: the Skp1-related proteins SKR-1/2, which are components of the Skp1, Cullin, F-box (SCF) ubiquitin ligase. Here, we find that the role of SKR-1 in the SC is conserved in Pristionchus pacificus. The P. pacificus Skp1 ortholog, Ppa-SKR-1, colocalizes with other SC proteins throughout meiotic prophase, where it occupies the middle of the SC. Like in C. elegans, the dimerization interface of Ppa-SKR-1 is required for its SC function. A dimerization mutant, ppa-skr-1F105E, fails to assemble SC and produces almost no progeny. Interestingly, the evolutionary trajectory of SKR-1 contrasts with other SC proteins. Unlike most SC proteins, SKR-1 is highly conserved in nematodes. Our results suggest that the structural role of SKR-1 in the SC has been conserved since the common ancestor of C. elegans and P. pacificus, and that rapidly evolving SC proteins have maintained the ability to interact with SKR-1 for at least 100 million years.
Collapse
Affiliation(s)
- Lisa E Kursel
- School of Biological Sciences, The University of Utah, Salt Lake City, UT 84112, USA
| | - Kaan Goktepe
- School of Biological Sciences, The University of Utah, Salt Lake City, UT 84112, USA
| | - Ofer Rog
- School of Biological Sciences, The University of Utah, Salt Lake City, UT 84112, USA
- Center for Cell and Genome Sciences, The University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
7
|
Olaya I, Burgess SM, Rog O. Formation and resolution of meiotic chromosome entanglements and interlocks. J Cell Sci 2024; 137:jcs262004. [PMID: 38985540 PMCID: PMC11267460 DOI: 10.1242/jcs.262004] [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: 07/12/2024] Open
Abstract
Interactions between parental chromosomes during the formation of gametes can lead to entanglements, entrapments and interlocks between unrelated chromosomes. If unresolved, these topological constraints can lead to misregulation of exchanges between chromosomes and to chromosome mis-segregation. Interestingly, these configurations are largely resolved by the time parental chromosomes are aligned during pachytene. In this Review, we highlight the inevitability of topologically complex configurations and discuss possible mechanisms to resolve them. We focus on the dynamic nature of a conserved chromosomal interface - the synaptonemal complex - and the chromosome movements that accompany meiosis as potential mechanisms to resolve topological constraints. We highlight the advantages of the nematode Caenorhabditis elegans for understanding biophysical features of the chromosome axis and synaptonemal complex that could contribute to mechanisms underlying interlock resolution. In addition, we highlight advantages of using the zebrafish, Danio rerio, as a model to understand how entanglements and interlocks are avoided and resolved.
Collapse
Affiliation(s)
- Iván Olaya
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA
- Integrative Genetics and Genomics Graduate Group, University of California Davis, Davis, CA 95616, USA
| | - Sean M. Burgess
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA
| | - Ofer Rog
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
Kursel LE, Goktepe K, Rog O. Skp1 is a conserved structural component of the meiotic synaptonemal complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600447. [PMID: 38979327 PMCID: PMC11230192 DOI: 10.1101/2024.06.24.600447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The synaptonemal complex (SC) is a meiotic interface that assembles between parental chromosomes and is essential for the formation of gametes. While the dimensions and ultrastructure of the SC are conserved across eukaryotes, its protein components are highly divergent. Recently, an unexpected component of the SC has been described in the nematode C. elegans: the Skp1-related proteins SKR-1/2, which are components of the Skp1, Cullin, F-box (SCF) ubiquitin ligase. Here, we find that the role of SKR-1 in the SC is conserved in nematodes. The P. pacificus Skp1 ortholog, Ppa-SKR-1, colocalizes with other SC proteins throughout meiotic prophase, where it occupies the middle of the SC. Like in C. elegans, the dimerization interface of Ppa-SKR-1 is required for its SC function. A dimerization mutant, Ppa-skr-1 F105E , fails to assemble SC and is almost completely sterile. Interestingly, the evolutionary trajectory of SKR-1 contrasts with other SC proteins. Unlike most SC proteins, SKR-1 is highly conserved in nematodes. Our results suggest that the structural role of SKR-1 in the SC has been conserved since the common ancestor of C. elegans and P. pacificus, and that rapidly evolving SC proteins have maintained the ability to interact with SKR-1 for at least 100 million years.
Collapse
Affiliation(s)
- Lisa E. Kursel
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, United States
| | - Kaan Goktepe
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, United States
| | - Ofer Rog
- School of Biological Sciences and Center for Cell and Genome Sciences, University of Utah, United States
| |
Collapse
|
9
|
Ishiguro KI. Mechanisms of meiosis initiation and meiotic prophase progression during spermatogenesis. Mol Aspects Med 2024; 97:101282. [PMID: 38797021 DOI: 10.1016/j.mam.2024.101282] [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: 02/17/2024] [Revised: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Meiosis is a critical step for spermatogenesis and oogenesis. Meiosis commences with pre-meiotic S phase that is subsequently followed by meiotic prophase. The meiotic prophase is characterized by the meiosis-specific chromosomal events such as chromosome recombination and homolog synapsis. Meiosis initiator (MEIOSIN) and stimulated by retinoic acid gene 8 (STRA8) initiate meiosis by activating the meiotic genes by installing the meiotic prophase program at pre-meiotic S phase. This review highlights the mechanisms of meiotic initiation and meiotic prophase progression from the point of the gene expression program and its relevance to infertility. Furthermore, upstream pathways that regulate meiotic initiation will be discussed in the context of spermatogenic development, indicating the sexual differences in the mode of meiotic entry.
Collapse
Affiliation(s)
- Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
10
|
Cossu IG, Leu NA, Guan Y, Wang PJ. The N-terminal modification of HORMAD2 causes its ectopic persistence on synapsed chromosomes without meiotic blockade. Reproduction 2024; 167:e230330. [PMID: 38401263 PMCID: PMC10993814 DOI: 10.1530/rep-23-0330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/22/2024] [Indexed: 02/26/2024]
Abstract
In brief The dissociation of HORMA domain protein 2 (HORMAD2) from the synaptonemal complex is tightly regulated. This study reveals that the N-terminal region of HORMAD2 is critical for its dissociation from synapsed meiotic chromosomes. Abstract During meiosis, homologous chromosomes undergo synapsis and recombination. HORMA domain proteins regulate key processes in meiosis. Mammalian HORMAD1 and HORMAD2 localize to unsynapsed chromosome axes but are removed upon synapsis by the TRIP13 AAA+ ATPase. TRIP13 engages the N-terminal region of HORMA domain proteins to induce an open conformation, resulting in the disassembly of protein complexes. Here, we report introduction of a 3×FLAG-HA tag to the N-terminus of HORMAD2 in mice. Coimmunoprecipitation coupled with mass spectrometry identified HORMAD1 and SYCP2 as HORMAD2-associated proteins in the testis. Unexpectedly, the N-terminal tagging of HORMAD2 resulted in its abnormal persistence along synapsed regions in pachynema and ectopic localization to telomeres in diplonema. Super-resolution microscopy revealed that 3×FLAG-HA-HORMAD2 was distributed along the central region of the synaptonemal complex, whereas wild-type HORMAD1 persisted along the lateral elements in 3×FLAG-HA-HORMAD2 meiocytes. Although homozygous mice completed meiosis and were fertile, homozygous males exhibited a significant reduction in sperm count. Collectively, these results suggest that the N-terminus of HORMAD2 is important for its timely removal from meiotic chromosome axes.
Collapse
Affiliation(s)
- Isabella G. Cossu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - N. Adrian Leu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Yongjuan Guan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
- College of Life Sciences, Capital Normal University, Beijing, China
| | - P. Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Zou M, Shabala S, Zhao C, Zhou M. Molecular mechanisms and regulation of recombination frequency and distribution in plants. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2024; 137:86. [PMID: 38512498 PMCID: PMC10957645 DOI: 10.1007/s00122-024-04590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
KEY MESSAGE Recent developments in understanding the distribution and distinctive features of recombination hotspots are reviewed and approaches are proposed to increase recombination frequency in coldspot regions. Recombination events during meiosis provide the foundation and premise for creating new varieties of crops. The frequency of recombination in different genomic regions differs across eukaryote species, with recombination generally occurring more frequently at the ends of chromosomes. In most crop species, recombination is rare in centromeric regions. If a desired gene variant is linked in repulsion with an undesired variant of a second gene in a region with a low recombination rate, obtaining a recombinant plant combining two favorable alleles will be challenging. Traditional crop breeding involves combining desirable genes from parental plants into offspring. Therefore, understanding the mechanisms of recombination and factors affecting the occurrence of meiotic recombination is important for crop breeding. Here, we review chromosome recombination types, recombination mechanisms, genes and proteins involved in the meiotic recombination process, recombination hotspots and their regulation systems and discuss how to increase recombination frequency in recombination coldspot regions.
Collapse
Affiliation(s)
- Meilin Zou
- Tasmanian Institute of Agriculture, University of Tasmania, Private Bag 1375, Prospect, TAS, 7250, Australia
| | - Sergey Shabala
- Tasmanian Institute of Agriculture, University of Tasmania, Private Bag 1375, Prospect, TAS, 7250, Australia
- School of Biological Sciences, University of Western Australia, 35 Stirling Highway, Perth, 6009, Australia
| | - Chenchen Zhao
- Tasmanian Institute of Agriculture, University of Tasmania, Private Bag 1375, Prospect, TAS, 7250, Australia
| | - Meixue Zhou
- Tasmanian Institute of Agriculture, University of Tasmania, Private Bag 1375, Prospect, TAS, 7250, Australia.
| |
Collapse
|
12
|
Vrielynck N, Peuch M, Durand S, Lian Q, Chambon A, Hurel A, Guérin J, Guérois R, Mercier R, Grelon M, Mézard C. SCEP1 and SCEP2 are two new components of the synaptonemal complex central element. NATURE PLANTS 2023; 9:2016-2030. [PMID: 37973938 DOI: 10.1038/s41477-023-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023]
Abstract
The synaptonemal complex (SC) is a proteinaceous structure that forms between homologous chromosomes during meiosis prophase. The SC is widely conserved across species, but its structure and roles during meiotic recombination are still debated. While the SC central region is made up of transverse filaments and central element proteins in mammals and fungi, few central element proteins have been identified in other species. Here we report the identification of two coiled-coil proteins, SCEP1 and SCEP2, that form a complex and localize at the centre of the Arabidopsis thaliana SC. In scep1 and scep2 mutants, chromosomes are aligned but not synapsed (the ZYP1 transverse filament protein is not loaded), crossovers are increased compared with the wild type, interference is lost and heterochiasmy is strongly reduced. We thus report the identification of two plant SC central elements, and homologues of these are found in all major angiosperm clades.
Collapse
Affiliation(s)
- Nathalie Vrielynck
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France
| | - Marion Peuch
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France
| | - Stéphanie Durand
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Qichao Lian
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Aurélie Chambon
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France
| | - Aurélie Hurel
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France
| | - Julie Guérin
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France
| | - Raphaël Guérois
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Raphaël Mercier
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Mathilde Grelon
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France.
| | - Christine Mézard
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin, Versailles, France.
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Institut Jean-Pierre Bourgin, Versailles, France.
| |
Collapse
|
13
|
Zhang Q, Zhang W, Wu X, Ke H, Qin Y, Zhao S, Guo T. Homozygous missense variant in MEIOSIN causes premature ovarian insufficiency. Hum Reprod 2023; 38:ii47-ii56. [PMID: 37982418 DOI: 10.1093/humrep/dead084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/30/2023] [Indexed: 11/21/2023] Open
Abstract
STUDY QUESTION Are variants of genes involved in meiosis initiation responsible for premature ovarian insufficiency (POI)? SUMMARY ANSWER A MEIOSIN variant participates in the pathogenesis of human POI by impairing meiosis due to insufficient transcriptional activation of essential meiotic genes. WHAT IS KNOWN ALREADY Meiosis is the key event for the establishment of the ovarian reserve, and several gene defects impairing meiotic homologous recombination have been found to contribute to the pathogenesis of POI. Although STRA8 and MEIOISN variants have been found to associate with POI in a recent study, the condition of other meiosis initiation genes is unknown and direct evidence of variants participating in the pathogenesis of POI is still lacking. STUDY DESIGN, SIZE, DURATION This was a retrospective genetic study. An in-house whole exome sequencing (WES) database of 1030 idiopathic POI patients was screened for variations of meiosis initiation genes. PARTICIPANTS/MATERIALS, SETTING, METHODS Homozygous or compound heterozygous variations of genes involved in meiosis initiation were screened in the in-house WES database. The pathogenicity of the variation was verified by in vitro experiments, including protein structure prediction and dual-luciferase reporter assay. The effect of the variant on ovarian function and meiosis was demonstrated through histological analyses in a point mutation mouse model. MAIN RESULTS AND THE ROLE OF CHANCE One homozygous variant in MEIOSIN (c.1735C>T, p.R579W) and one in STRA8 (c.258 + 1G>A), which initiates meiosis via the retinoic acid-dependent pathway, were identified in a patient with idiopathic POI respectively. The STRA8 variation has been reported in the recently published work. For the MEIOSIN variation, the dual-luciferase reporter assay revealed that the variant adversely affected the transcriptional function of MEIOSIN in upregulating meiotic genes. Furthermore, knock-in mice with the homologous mutation confirmed that the variation impacted the meiotic prophase I program and accelerated oocyte depletion. Moreover, the variant p.R579W localizing in the high-mobility group (HMG) box domain disrupted the nuclear localization of the MEIOSIN protein but was dispensable for the cell-cycle switch of oocytes, suggesting a unique role of the MEIOSIN HMG box domain in meiosis initiation. LIMITATIONS, REASONS FOR CAUTION Further studies are needed to explore the role of other meiosis initiation genes in the pathogenesis of POI. WIDER IMPLICATIONS OF THE FINDINGS The MEIOSIN variant was verified to cause POI by impaired transcriptional regulation of meiotic genes and was inherited by a recessive mode. The function of HMG box domain in MEIOSIN protein was also expanded by this study. Although causative variations in meiotic initiation genes are rare in POI, our study confirmed the pathogenicity of a MEIOSIN variant and elucidated another mechanism of human infertility. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Key Research & Developmental Program of China (2022YFC2703800, 2022YFC2703000), National Natural Science Foundation for Distinguished Young Scholars (82125014), National Natural Science Foundation of China (32070847, 32170867, 82071609), Basic Science Center Program of NSFC (31988101), Natural Science Foundation of Shandong Province for Grand Basic Projects (ZR2021ZD33), Natural Science Foundation of Shandong Province for Excellent Young Scholars (ZR2022YQ69), Taishan Scholars Program for Young Experts of Shandong Province (tsqn202211371), and Qilu Young Scholars Program of Shandong University. The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Qian Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Wenzhe Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Xinyi Wu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Hanni Ke
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Shidou Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| | - Ting Guo
- Center for Reproductive Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
| |
Collapse
|
14
|
Billmyre KK, Kesler EA, Tsuchiya D, Corbin TJ, Weaver K, Moran A, Yu Z, Adams L, Delventhal K, Durnin M, Davies OR, Hawley RS. SYCP1 head-to-head assembly is required for chromosome synapsis in mouse meiosis. SCIENCE ADVANCES 2023; 9:eadi1562. [PMID: 37862414 PMCID: PMC10588951 DOI: 10.1126/sciadv.adi1562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/20/2023] [Indexed: 10/22/2023]
Abstract
In almost all sexually reproducing organisms, meiotic recombination and cell division require the synapsis of homologous chromosomes by a large proteinaceous structure, the synaptonemal complex (SC). While the SC's overall structure is highly conserved across eukaryotes, its constituent proteins diverge between phyla. Transverse filament protein, SYCP1, spans the width of the SC and undergoes amino-terminal head-to-head self-assembly in vitro through a motif that is unusually highly conserved across kingdoms of life. Here, we report creation of mouse mutants, Sycp1L102E and Sycp1L106E, that target SYCP1's head-to-head interface. L106E resulted in a complete loss of synapsis, while L102E had no apparent effect on synapsis, in agreement with their differential effects on the SYCP1 head-to-head interface in molecular dynamics simulations. In Sycp1L106E mice, homologs aligned and recruited low levels of mutant SYCP1 and other SC proteins, but the absence of synapsis led to failure of crossover formation and meiotic arrest. We conclude that SYCP1's conserved head-to-head interface is essential for meiotic chromosome synapsis in vivo.
Collapse
Affiliation(s)
| | - Emily A. Kesler
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Kyle Weaver
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Andrea Moran
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Lane Adams
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Kym Delventhal
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Michael Durnin
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Owen Richard Davies
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - R. Scott Hawley
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
15
|
Chi RPA, Xu X, Li JL, Xu X, Hu G, Brown P, Willson C, Kirsanov O, Geyer C, Huang CL, Morgan M, DeMayo F. WNK1 is required during male pachynema to sustain fertility. iScience 2023; 26:107616. [PMID: 37694147 PMCID: PMC10485039 DOI: 10.1016/j.isci.2023.107616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/04/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
WNK1 is an important regulator in many physiological functions, yet its role in male reproduction is unexplored. In the male germline, WNK1 is upregulated in preleptotene spermatocytes indicating possible function(s) in spermatogenic meiosis. Indeed, deletion of Wnk1 in mid-pachytene spermatocytes using the Wnt7a-Cre mouse led to male sterility which resembled non-obstructive azoospermia in humans, where germ cells failed to complete spermatogenesis and produced no sperm. Mechanistically, we found elevated MTOR expression and signaling in the Wnk1-depleted spermatocytes. As MTOR is a central mediator of translation, we speculated that translation may be accelerated in these spermatocytes. Supporting this, we found the acrosome protein, ACRBP to be prematurely expressed in the spermatocytes with Wnk1 deletion. Our study uncovered an MTOR-regulating factor in the male germline with potential implications in translation, and future studies will aim to understand how WNK1 regulates MTOR activity and impact translation on a broader spectrum.
Collapse
Affiliation(s)
- Ru-pin Alicia Chi
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Xin Xu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Paula Brown
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Cynthia Willson
- Integrated Laboratory Systems LLC, Research Triangle Park, NC 27709, USA
| | - Oleksandr Kirsanov
- Department of Anatomy & Cell Biology at the Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Christopher Geyer
- Department of Anatomy & Cell Biology at the Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Chou-Long Huang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa, IA 52242, USA
| | - Marcos Morgan
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Francesco DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| |
Collapse
|
16
|
Rosa IR, Barbosa CP, Ferrandez CA, Sonoda BDB, Christofolini DM, Trevisan CM, Laganà AS, Peluso C, Bianco B. Involvement of SYCP2L and TDRD3 gene variants on ovarian reserve and reproductive outcomes: a cross-sectional study. JBRA Assist Reprod 2023; 27:428-435. [PMID: 37417852 PMCID: PMC10712833 DOI: 10.5935/1518-0557.20220074] [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: 06/21/2022] [Accepted: 12/26/2022] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVE Single nucleotide variants have been implicated in the response to fertility treatment and a pharmacogenomic approach may help to customize therapy based on patient genome. We aimed to evaluate the effect, individual and combined, of SYCP2L (rs2153157:G>A) and TDRD3 (rs4886238:G>A) variants on ovarian reserve, response to controlled ovarian stimulation (COS) and reproductive outcomes of women undergoing in vitro fertilization (IVF) treatment. METHODS This cross-sectional study included 149 normoovulatory women undergoing IVF. Genotyping was performed using the TaqMan real-time polymerase chain reaction method. Clinical parameters and reproductive outcomes were compared according to the genotypes of the variants studied. RESULTS Considering ovarian reserve, there were no significant differences among SYCP2L or TDRD3 genotypes in terms of FSH levels or AFC; however, AMH levels were significantly different in carriers of both variants. Regarding the SYCP2L rs2153157:G>A variant, lower AMH levels were observed in women carrying an AA genotype compared to women carrying a heterozygous genotype (p=0.01). Considering the TDRD3 rs4886238:G>A variant, women carrying an AA genotype presented higher AMH levels than carriers of GG and GA genotypes (p=0.025). Nevertheless, no difference was found regarding response to COS or reproductive outcomes. Considering the combined effect of the variants, women carrying the heterozygous genotype of both variants presented statistically increased AMH levels compared to SYCP2L rs2153157 AA genotype carriers and TDRD3 rs4886238 GG genotype carriers (p=0.042). CONCLUSIONS Individually and combined, the SYCP2L rs2153157 and TDRD3 rs4886238 variants have an effect on AMH level.
Collapse
Affiliation(s)
- Iasmim Ribeiro Rosa
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Caio Parente Barbosa
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| | - Caroline Awoki Ferrandez
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Bianca Del Bel Sonoda
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Denise Maria Christofolini
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| | - Camila Martins Trevisan
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS “Civico - Di Cristina -
Benfratelli”, Department of Health Promotion, Mother and Child Care, Internal
Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Carla Peluso
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Bianca Bianco
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| |
Collapse
|
17
|
Bui MD, Luong TLA, Tran HD, Duong TTH, Nguyen TN, Nguyen DT, Nguyen TD, Nong VH. A Novel Frameshift Microdeletion of the TEX12 Gene Caused Infertility in Two Brothers with Nonobstructive Azoospermia. Reprod Sci 2023; 30:2876-2881. [PMID: 37012491 DOI: 10.1007/s43032-023-01226-8] [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: 12/23/2022] [Accepted: 03/27/2023] [Indexed: 04/05/2023]
Abstract
Male infertility is a growing health problem, which affects approximately 7% of the global male population. Nonobstructive azoospermia (NOA) is one of the most severe forms of male infertility caused by genetic defects, including chromosome structural abnormalities, Y chromosome microdeletions, or single-gene alterations. However, the etiology of up to 40% of NOA cases is unidentified. By whole-exome sequencing, we detected a homozygous 5-bp-deletion variant in exon 4 of the TEX12 gene (c.196-200del, p.L66fs, NM_031275.4) in two brothers with NOA of a nonconsanguineous Vietnamese family. This deletion variant of 5 nucleotides (ATTAG) results in a premature stop codon in exon 4 and truncation of the C-terminal. Segregation analysis by Sanger sequencing confirmed that the deletion variant was inherited in an autosomal recessive pattern. The 1st and 3rd infertile sons were homozygous for the deletion, whereas the 2nd fertile son and both parents were heterozygous. The new deletion mutation identified in TEX12 gene caused loss of function of TEX12 gene. The loss of TEX12 function has already caused infertility in male mice. Therefore, we concluded that the loss of TEX12 function may cause infertility in men. To our knowledge, this is the first case reported so far indicating disruption of human TEX12, which leads to infertility in men.
Collapse
Affiliation(s)
- Minh Duc Bui
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | - Huu Dinh Tran
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Thi Thu Ha Duong
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Thy Ngoc Nguyen
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Dang Ton Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Thuy Duong Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.
| | - Van Hai Nong
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.
| |
Collapse
|
18
|
Abstract
In meiosis, homologous chromosome synapsis is mediated by a supramolecular protein structure, the synaptonemal complex (SC), that assembles between homologous chromosome axes. The mammalian SC comprises at least eight largely coiled-coil proteins that interact and self-assemble to generate a long, zipper-like structure that holds homologous chromosomes in close proximity and promotes the formation of genetic crossovers and accurate meiotic chromosome segregation. In recent years, numerous mutations in human SC genes have been associated with different types of male and female infertility. Here, we integrate structural information on the human SC with mouse and human genetics to describe the molecular mechanisms by which SC mutations can result in human infertility. We outline certain themes in which different SC proteins are susceptible to different types of disease mutation and how genetic variants with seemingly minor effects on SC proteins may act as dominant-negative mutations in which the heterozygous state is pathogenic.
Collapse
Affiliation(s)
- Ian R Adams
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom;
| | - Owen R Davies
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom;
| |
Collapse
|
19
|
Llano E, Pendás AM. Synaptonemal Complex in Human Biology and Disease. Cells 2023; 12:1718. [PMID: 37443752 PMCID: PMC10341275 DOI: 10.3390/cells12131718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
The synaptonemal complex (SC) is a meiosis-specific multiprotein complex that forms between homologous chromosomes during prophase of meiosis I. Upon assembly, the SC mediates the synapses of the homologous chromosomes, leading to the formation of bivalents, and physically supports the formation of programmed double-strand breaks (DSBs) and their subsequent repair and maturation into crossovers (COs), which are essential for genome haploidization. Defects in the assembly of the SC or in the function of the associated meiotic recombination machinery can lead to meiotic arrest and human infertility. The majority of proteins and complexes involved in these processes are exclusively expressed during meiosis or harbor meiosis-specific subunits, although some have dual functions in somatic DNA repair and meiosis. Consistent with their functions, aberrant expression and malfunctioning of these genes have been associated with cancer development. In this review, we focus on the significance of the SC and their meiotic-associated proteins in human fertility, as well as how human genetic variants encoding for these proteins affect the meiotic process and contribute to infertility and cancer development.
Collapse
Affiliation(s)
- Elena Llano
- Departamento Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biologıía Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain;
| | - Alberto M. Pendás
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biologıía Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain;
| |
Collapse
|
20
|
Ozturk S. Genetic variants underlying spermatogenic arrests in men with non-obstructive azoospermia. Cell Cycle 2023; 22:1021-1061. [PMID: 36740861 PMCID: PMC10081088 DOI: 10.1080/15384101.2023.2171544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 02/07/2023] Open
Abstract
Spermatogenic arrest is a severe form of non-obstructive azoospermia (NOA), which occurs in 10-15% of infertile men. Interruption in spermatogenic progression at premeiotic, meiotic, or postmeiotic stage can lead to arrest in men with NOA. Recent studies have intensively focused on defining genetic variants underlying these spermatogenic arrests by making genome/exome sequencing. A number of variants were discovered in the genes involving in mitosis, meiosis, germline differentiation and other basic cellular events. Herein, defined variants in NOA cases with spermatogenic arrests and created knockout mouse models for the related genes are comprehensively reviewed. Also, importance of gene panel-based screening for NOA cases was discussed. Screening common variants in these infertile men with spermatogenic arrests may contribute to elucidating the molecular background and designing novel treatment strategies.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
21
|
Herrera LR, McGlynn K, Gibbs ZA, Davis AJ, Whitehurst AW. The Cancer Testes Antigen, HORMAD1, is a Tumor-Specific Replication Fork Protection Factor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526348. [PMID: 36778501 PMCID: PMC9915569 DOI: 10.1101/2023.01.31.526348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumors frequently activate the expression of genes that are only otherwise required for meiosis. HORMAD1, which is essential for meiotic recombination in multiple species, is expressed in over 50% of human lung adenocarcinoma cells (LUAD). We previously found that HORMAD1 promotes DNA double strand break (DSB) repair in LUAD. Here, we report that HORMAD1 takes on an additional role in protecting genomic integrity. Specifically, we find HORMAD1 is critical for protecting stalled DNA replication forks in LUAD. Loss of HORMAD1 leads to nascent DNA degradation, an event which is mediated by the MRE11-DNA2-BLM pathway. Moreover, following exogenous induction of DNA replication stress, HORMAD1 deleted cells accumulate single stranded DNA (ssDNA). We find that these phenotypes are the result of a lack of RAD51 and BRCA2 loading onto stalled replication forks. Ultimately, loss of HORMAD1 leads to increased DSBs and chromosomal aberrations in response to replication stress. Collectively, our data support a model where HORMAD1 expression is selected to mitigate DNA replication stress, which would otherwise induce deleterious genomic instability.
Collapse
|
22
|
Crichton JH, Dunce JM, Dunne OM, Salmon LJ, Devenney PS, Lawson J, Adams IR, Davies OR. Structural maturation of SYCP1-mediated meiotic chromosome synapsis by SYCE3. Nat Struct Mol Biol 2023; 30:188-199. [PMID: 36635604 PMCID: PMC7614228 DOI: 10.1038/s41594-022-00909-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/06/2022] [Indexed: 01/13/2023]
Abstract
In meiosis, a supramolecular protein structure, the synaptonemal complex (SC), assembles between homologous chromosomes to facilitate their recombination. Mammalian SC formation is thought to involve hierarchical zipper-like assembly of an SYCP1 protein lattice that recruits stabilizing central element (CE) proteins as it extends. Here we combine biochemical approaches with separation-of-function mutagenesis in mice to show that, rather than stabilizing the SYCP1 lattice, the CE protein SYCE3 actively remodels this structure during synapsis. We find that SYCP1 tetramers undergo conformational change into 2:1 heterotrimers on SYCE3 binding, removing their assembly interfaces and disrupting the SYCP1 lattice. SYCE3 then establishes a new lattice by its self-assembly mimicking the role of the disrupted interface in tethering together SYCP1 dimers. SYCE3 also interacts with CE complexes SYCE1-SIX6OS1 and SYCE2-TEX12, providing a mechanism for their recruitment. Thus, SYCE3 remodels the SYCP1 lattice into a CE-binding integrated SYCP1-SYCE3 lattice to achieve long-range synapsis by a mature SC.
Collapse
Affiliation(s)
- James H Crichton
- MRC Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - James M Dunce
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Orla M Dunne
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Vienna BioCenter Core Facilities GmbH, Vienna, Austria
| | - Lucy J Salmon
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Paul S Devenney
- MRC Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jennifer Lawson
- MRC Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ian R Adams
- MRC Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | - Owen R Davies
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
23
|
Functions of Steroid Hormones in the Male Reproductive Tract as Revealed by Mouse Models. Int J Mol Sci 2023; 24:ijms24032748. [PMID: 36769069 PMCID: PMC9917565 DOI: 10.3390/ijms24032748] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Steroid hormones are capable of diffusing through cell membranes to bind with intracellular receptors to regulate numerous physiological processes. Three classes of steroid hormones, namely androgens, estrogens and glucocorticoids, contribute to the development of the reproductive system and the maintenance of fertility. During the past 30 years, mouse models have been produced in which the expression of genes encoding steroid hormone receptors has been enhanced, partially compromised or eliminated. These mouse models have revealed many of the physiological processes regulated by androgens, estrogens and to a more limited extent glucocorticoids in the testis and male accessory organs. In this review, advances provided by mouse models that have facilitated a better understanding of the molecular regulation of testis and reproductive tract processes by steroid hormones are discussed.
Collapse
|
24
|
Ito M, Shinohara A. Chromosome architecture and homologous recombination in meiosis. Front Cell Dev Biol 2023; 10:1097446. [PMID: 36684419 PMCID: PMC9853400 DOI: 10.3389/fcell.2022.1097446] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023] Open
Abstract
Meiocytes organize higher-order chromosome structures comprising arrays of chromatin loops organized at their bases by linear axes. As meiotic prophase progresses, the axes of homologous chromosomes align and synapse along their lengths to form ladder-like structures called synaptonemal complexes (SCs). The entire process of meiotic recombination, from initiation via programmed DNA double-strand breaks (DSBs) to completion of DSB repair with crossover or non-crossover outcomes, occurs in the context of chromosome axes and SCs. These meiosis-specific chromosome structures provide specialized environments for the regulation of DSB formation and crossing over. In this review, we summarize insights into the importance of chromosome architecture in the regulation of meiotic recombination, focusing on cohesin-mediated axis formation, DSB regulation via tethered loop-axis complexes, inter-homolog template bias facilitated by axial proteins, and crossover regulation in the context of the SCs. We also discuss emerging evidence that the SUMO and the ubiquitin-proteasome system function in the organization of chromosome structure and regulation of meiotic recombination.
Collapse
Affiliation(s)
- Masaru Ito
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Akira Shinohara
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
25
|
Huang Y, Roig I. Genetic control of meiosis surveillance mechanisms in mammals. Front Cell Dev Biol 2023; 11:1127440. [PMID: 36910159 PMCID: PMC9996228 DOI: 10.3389/fcell.2023.1127440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Meiosis is a specialized cell division that generates haploid gametes and is critical for successful sexual reproduction. During the extended meiotic prophase I, homologous chromosomes progressively pair, synapse and desynapse. These chromosomal dynamics are tightly integrated with meiotic recombination (MR), during which programmed DNA double-strand breaks (DSBs) are formed and subsequently repaired. Consequently, parental chromosome arms reciprocally exchange, ultimately ensuring accurate homolog segregation and genetic diversity in the offspring. Surveillance mechanisms carefully monitor the MR and homologous chromosome synapsis during meiotic prophase I to avoid producing aberrant chromosomes and defective gametes. Errors in these critical processes would lead to aneuploidy and/or genetic instability. Studies of mutation in mouse models, coupled with advances in genomic technologies, lead us to more clearly understand how meiosis is controlled and how meiotic errors are linked to mammalian infertility. Here, we review the genetic regulations of these major meiotic events in mice and highlight our current understanding of their surveillance mechanisms. Furthermore, we summarize meiotic prophase genes, the mutations that activate the surveillance system leading to meiotic prophase arrest in mouse models, and their corresponding genetic variants identified in human infertile patients. Finally, we discuss their value for the diagnosis of causes of meiosis-based infertility in humans.
Collapse
Affiliation(s)
- Yan Huang
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ignasi Roig
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
26
|
A cryo-fixation protocol to study the structure of the synaptonemal complex. Chromosome Res 2022; 30:385-400. [PMID: 35486207 DOI: 10.1007/s10577-022-09689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/28/2022] [Indexed: 01/25/2023]
Abstract
Genetic variability in sexually reproducing organisms results from an exchange of genetic material between homologous chromosomes. The genetic exchange mechanism is dependent on the synaptonemal complex (SC), a protein structure localized between the homologous chromosomes. The current structural models of the mammalian SC are based on electron microscopy, superresolution, and expansion microscopy studies using chemical fixatives and sample dehydration of gonads, which are methodologies known to produce structural artifacts. To further analyze the structure of the SC, without chemical fixation, we have adapted a cryo-fixation method for electron microscopy where pachytene cells are isolated from mouse testis by FACS, followed by cryo-fixation, cryo-substitution, and electron tomography. In parallel, we performed conventional chemical fixation and electron tomography on mouse seminiferous tubules to compare the SC structure obtained with the two fixation methods. We found several differences in the structure and organization of the SC in cryo-fixed samples when compared to chemically preserved samples. We found the central region of the SC to be wider and the transverse filaments to be more densely packed in the central region of the SC.
Collapse
|
27
|
Wyrwoll MJ, Wabschke R, Röpke A, Wöste M, Ruckert C, Perrey S, Rotte N, Hardy J, Astica L, Lupiáñez DG, Wistuba J, Westernströer B, Schlatt S, Berman AJ, Müller AM, Kliesch S, Yatsenko AN, Tüttelmann F, Friedrich C. Analysis of copy number variation in men with non-obstructive azoospermia. Andrology 2022; 10:1593-1604. [PMID: 36041235 PMCID: PMC9605881 DOI: 10.1111/andr.13267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Recent findings demonstrate that single nucleotide variants can cause non-obstructive azoospermia (NOA). In contrast, copy number variants (CNVs) were only analysed in few studies in infertile men. Some have reported a higher prevalence of CNVs in infertile versus fertile men. OBJECTIVES This study aimed to elucidate if CNVs are associated with NOA. MATERIALS AND METHODS We performed array-based comparative genomic hybridisation (aCGH) in 37 men with meiotic arrest, 194 men with Sertoli cell-only phenotype, and 21 control men. We filtered our data for deletions affecting genes and prioritised the affected genes according to the literature search. Prevalence of CNVs was compared between all groups. Exome data of 2,030 men were screened to detect further genetic variants in prioritised genes. Modelling was performed for the protein encoded by the novel candidate gene TEKT5 and we stained for TEKT5 in human testicular tissue. RESULTS We determined the cause of infertility in two individuals with homozygous deletions of SYCE1 and in one individual with a heterozygous deletion of SYCE1 combined with a likely pathogenic missense variant on the second allele. We detected heterozygous deletions affecting MLH3, EIF2B2, SLX4, CLPP and TEKT5, in one subject each. CNVs were not detected more frequently in infertile men compared with controls. DISCUSSION While SYCE1 and MLH3 encode known meiosis-specific proteins, much less is known about the proteins encoded by the other identified candidate genes, warranting further analyses. We were able to identify the cause of infertility in one out of the 231 infertile men by aCGH and in two men by using exome sequencing data. CONCLUSION As aCGH and exome sequencing are both expensive methods, combining both in a clinical routine is not an effective strategy. Instead, using CNV calling from exome data has recently become more precise, potentially making aCGH dispensable.
Collapse
Affiliation(s)
- M. J. Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - R. Wabschke
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - A. Röpke
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - M. Wöste
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - C. Ruckert
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - S. Perrey
- Institute for Bioinformatics and Chemoinformatics, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - N. Rotte
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - J. Hardy
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - L. Astica
- Epigenetics and Sex Development Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - D. G. Lupiáñez
- Epigenetics and Sex Development Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - J. Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - B. Westernströer
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - S. Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - A. J. Berman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - A. M. Müller
- Practice for Pathology and Centre for Pediatric Pathology, University Hospital of Cologne, Cologne, Germany
| | - S. Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - A. N. Yatsenko
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - F. Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - C. Friedrich
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| |
Collapse
|
28
|
Wang Q, Yan Q, Nan J, Wang J, Zhang Y, Zhao X. Syce1 and Syce3 regulate testosterone and dihydrotestosterone synthesis via steroidogenic pathways in mouse Sertoli and Leydig cells. J Steroid Biochem Mol Biol 2022; 223:106135. [PMID: 35697131 DOI: 10.1016/j.jsbmb.2022.106135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 11/28/2022]
Abstract
Testosterone (T) and dihydrotestosterone (DHT) are the main hormones regulating reproduction and development of male animals. Although their synthesis and secretion are regulated by the endocrine system [hypothalamic-pituitary-gonadal (adrenal) axis], it is also possible to synthesize T and DHT from the induction of two proteins: Syce1 and Syce3. As central elements of the synaptonemal complex (SC), Syce1 and Syce3 play a key role in the association of homologous chromosomes during meiosis. However, Syce1 and Syce3 also promote the synthesis of T and DHT, although potential mechanisms have yet to be revealed. In this study, Leydig and Sertoli cells, which are responsible for the production and regulation of steroid hormones in testis, were transfected with recombinant Syce1/Syce3 and silence sequence. Our results revealed the highest expression of Syce1 and Syce3 in spermatogenic cells of the testis. Moreover, overexpression or knockdown of Syce1 and Syce3 in Sertoli and Leydig cells resulted in activation or suppression of steroidogenic genes Star and Hsd3b, which are involved in a steroidogenic pathway that upregulates T synthesis. Upregulated expression of Syce1 resulted in a significant increase in Srd5a1, which can promote DHT secretion. Interestingly, Syce1 and Syce3 overexpression synergistically promoted each other's abundance. Our results define a previously unknown mechanism of Syce1 and Syce3 dependent activation of steroidogenic signaling in Sertoli and Leydig cells.
Collapse
Affiliation(s)
- Qi Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
| | - Qiu Yan
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
| | - Jinghong Nan
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
| | - Jie Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China.
| |
Collapse
|
29
|
Luo H, Mipam T, Wu S, Xu C, Yi C, Zhao W, Chai Z, Chen X, Wu Z, Wang J, Wang J, Wang H, Zhong J, Cai X. DNA methylome of primary spermatocyte reveals epigenetic dysregulation associated with male sterility of cattleyak. Theriogenology 2022; 191:153-167. [PMID: 35988507 DOI: 10.1016/j.theriogenology.2022.08.016] [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: 10/01/2021] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
DNA cytosine methylation modification in the germline is of particular importance since it is a highly heritable epigenetic mark. Although cytosine methylation has been analyzed at the genome-scale for several mammalian species, our knowledge of DNA methylation patterns and the mechanisms underlying male hybrid sterility is still limited in domestic animals such as cattleyak. Here we for the first time show the genome-wide and single-base resolution landscape of methylcytosines (mC) in the primary spermatocyte (PSC) genome of yak with normal spermatogenesis and the inter-specific hybrid cattleyak with male infertility. A comparative investigation revealed that widespread differences are observed in the composition and patterning of DNA cytosine methylation between the two methylomes. Global CG or non-CG DNA methylation levels, as well as the number of mC sites, are increased in cattleyak compared to yak. Notably, the DNA methylome in cattleyak PSC exhibits promoter hypermethylation of meiosis-specific genes and piRNA pathway genes with respect to yak. Furthermore, major retrotransposonson classes are predominantly hypermethylated in cattleyak while those are fully hypomethylated in yak. KEGG pathway enrichment indicates Rap1 signaling and MAPK pathways may play potential roles in the spermatogenic arrest of cattleyak. Our present study not only provides valuable insights into distinct features of the cattleyak PSC methylome but also paves the way toward elucidating the complex, yet highly coordinated epigenetic modification during male germline development for inter-specific hybrid animals.
Collapse
Affiliation(s)
- Hui Luo
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - TserangDonko Mipam
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Shixin Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Chuanfei Xu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Chuanping Yi
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Xuemei Chen
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Zhijuan Wu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Jikun Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Jiabo Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China.
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
30
|
PRC1-mediated epigenetic programming is required to generate the ovarian reserve. Nat Commun 2022; 13:4510. [PMID: 35948547 PMCID: PMC9365831 DOI: 10.1038/s41467-022-31759-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022] Open
Abstract
The ovarian reserve defines the female reproductive lifespan, which in humans spans decades due to robust maintenance of meiotic arrest in oocytes residing in primordial follicles. Epigenetic reprogramming, including DNA demethylation, accompanies meiotic entry, but the chromatin changes that underpin the generation and preservation of ovarian reserves are poorly defined. We report that the Polycomb Repressive Complex 1 (PRC1) establishes repressive chromatin states in perinatal mouse oocytes that directly suppress the gene expression program of meiotic prophase-I and thereby enable the transition to dictyate arrest. PRC1 dysfuction causes depletion of the ovarian reserve and leads to premature ovarian failure. Our study demonstrates a fundamental role for PRC1-mediated gene silencing in female reproductive lifespan, and reveals a critical window of epigenetic programming required to establish ovarian reserve. In humans, the ovarian reserve is maintained over decades by meiotic arrest of oocytes. Here the authors show that Polycomb Repressive Complex 1 (PRC1)-mediated epigenetic programming is essential for formation of ovarian reserve and thus female reproductive lifespan.
Collapse
|
31
|
Mouse oocytes carrying metacentric Robertsonian chromosomes have fewer crossover sites and higher aneuploidy rates than oocytes carrying acrocentric chromosomes alone. Sci Rep 2022; 12:12028. [PMID: 35835815 PMCID: PMC9283534 DOI: 10.1038/s41598-022-16175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 07/06/2022] [Indexed: 12/03/2022] Open
Abstract
Meiotic homologous recombination during fetal development dictates proper chromosome segregation in adult mammalian oocytes. Successful homologous synapsis and recombination during Meiotic Prophase I (MPI) depends on telomere-led chromosome movement along the nuclear envelope. In mice, all chromosomes are acrocentric, while other mammalian species carry a mixture of acrocentric and metacentric chromosomes. Such differences in telomeric structures may explain the exceptionally low aneuploidy rates in mice. Here, we tested whether the presence of metacentric chromosomes carrying Robertsonian translocations (RbT) affects the rate of homologous recombination or aneuploidy. We found a delay in MPI progression in RbT-carrier vs. wild-type (WT) fetal ovaries. Furthermore, resolution of distal telomere clusters, associated with synapsis initiation, was delayed and centromeric telomere clusters persisted until later MPI substages in RbT-carrier oocytes compared to WT oocytes. When chromosomes fully synapsed, higher percentages of RbT-carrier oocytes harbored at least one chromosome pair lacking MLH1 foci, which indicate crossover sites, compared to WT oocytes. Aneuploidy rates in ovulated eggs were also higher in RbT-carrier females than in WT females. In conclusion, the presence of metacentric chromosomes among acrocentric chromosomes in mouse oocytes delays MPI progression and reduces the efficiency of homologous crossover, resulting in a higher frequency of aneuploidy.
Collapse
|
32
|
TCFL5 deficiency impairs the pachytene to diplotene transition during spermatogenesis in the mouse. Sci Rep 2022; 12:10956. [PMID: 35768632 PMCID: PMC9242989 DOI: 10.1038/s41598-022-15167-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/20/2022] [Indexed: 11/27/2022] Open
Abstract
Spermatogenesis is a complex, multistep process during which spermatogonia give rise to spermatozoa. Transcription Factor Like 5 (TCFL5) is a transcription factor that has been described expressed during spermatogenesis. In order to decipher the role of TCFL5 during in vivo spermatogenesis, we generated two mouse models. Ubiquitous removal of TCFL5 generated by breeding TCFL5fl/fl with SOX2-Cre mice resulted in sterile males being unable to produce spermatozoa due to a dramatic alteration of the testis architecture presenting meiosis arrest and lack of spermatids. SYCP3, SYCP1 and H1T expression analysis showed that TCFL5 deficiency causes alterations during pachytene/diplotene transition resulting in a meiotic arrest in a diplotene-like stage. Even more, TCFL5 deficient pachytene showed alterations in the number of MLH1 foci and the condensation of the sexual body. In addition, tamoxifen-inducible TCFL5 knockout mice showed, besides meiosis phenotype, alterations in the spermatids elongation process resulting in aberrant spermatids. Furthermore, TCFL5 deficiency increased spermatogonia maintenance genes (Dalz, Sox2, and Dmrt1) but also increased meiosis genes (Syce1, Stag3, and Morc2a) suggesting that the synaptonemal complex forms well, but cannot separate and meiosis does not proceed. TCFL5 is able to bind to the promoter of Syce1, Stag3, Dmrt1, and Syce1 suggesting a direct control of their expression. In conclusion, TCFL5 plays an essential role in spermatogenesis progression being indispensable for meiosis resolution and spermatids maturation.
Collapse
|
33
|
Steinberg ER, Bressa MJ, Mudry MD. Sex chromosome systems in Neotropical Primates: What have we learnt so far from cytogenetics and genomics? J Evol Biol 2022; 35:1589-1600. [PMID: 35731796 DOI: 10.1111/jeb.14039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 11/29/2022]
Abstract
Neotropical Primates (Platyrrhini) show great diversity in their life histories, ecology, behaviour and genetics. This diversity extends to their chromosome complements, both to autosomes and to sex chromosomes. In this contribution, we will review what is currently known about sex chromosomes in this group, both from cytogenetic and from genomic evidence. The X and Y chromosomes in Neotropical Primates, also known as New World Monkeys, have striking structural differences compared with Old World Monkeys when Catarrhini sex chromosomes are considered. The XY bivalent displays a different meiotic behaviour in prophase I, and their Y chromosome shows extensive genomic differences. Even though the most widespread sex chromosome system is the XX/XY and thus considered the ancestral one for Platyrrhini, modifications of this sexual system are observed within this group. Multiple sex chromosome systems originated from Y-autosome translocations were described in several genera (Aotus, Callimico and Alouatta). In the howler monkeys, genus Alouatta, an independent origin of the sexual systems in South American and Mesoamerican species was postulated. All the above-mentioned evidence suggests that the Y chromosome of Platyrrhini has a different evolutionary history compared with the Catarrhini Y. There is still much to understand regarding their sex chromosome systems.
Collapse
Affiliation(s)
- Eliana Ruth Steinberg
- Grupo de Investigación en Biología Evolutiva (GIBE). Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Departamento de Ecología, Genética y Evolución (EGE), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), CONICET, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María José Bressa
- Grupo de Citogenética de Insectos. Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Departamento de Ecología, Genética y Evolución (EGE), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), CONICET, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Marta Dolores Mudry
- Grupo de Investigación en Biología Evolutiva (GIBE). Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA), Departamento de Ecología, Genética y Evolución (EGE), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), CONICET, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
34
|
Abstract
Meiosis is critical for germ cell development in multicellular organisms. Initiation of meiosis coincides with pre-meiotic S phase, which is followed by meiotic prophase, a prolonged G2 phase that ensures numerous meiosis-specific chromosome events. Meiotic prophase is accompanied by robust alterations of gene expression. In mouse germ cells, MEIOSIN and STRA8 direct cell cycle switch from mitosis to meiosis. MEIOSIN and STRA8 coordinate meiotic initiation with cell cycle, by activating the meiotic genes to have meiotic prophase program installed at S phase. This review mainly focuses on the mechanism of meiotic initiation in mouse germ cells from the viewpoint of the transcription of meiotic genes. Furthermore, signaling pathways that regulate meiotic initiation will be discussed in the context of germ cell development, pointing out the sexual differences in the mode of meiotic initiation.
Collapse
Affiliation(s)
- Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
35
|
Ravindranathan R, Raveendran K, Papanikos F, San-Segundo P, Tóth A. Chromosomal synapsis defects can trigger oocyte apoptosis without elevating numbers of persistent DNA breaks above wild-type levels. Nucleic Acids Res 2022; 50:5617-5634. [PMID: 35580048 PMCID: PMC9177993 DOI: 10.1093/nar/gkac355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/08/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
Generation of haploid gametes depends on a modified version of homologous recombination in meiosis. Meiotic recombination is initiated by single-stranded DNA (ssDNA) ends originating from programmed DNA double-stranded breaks (DSBs) that are generated by the topoisomerase-related SPO11 enzyme. Meiotic recombination involves chromosomal synapsis, which enhances recombination-mediated DSB repair, and thus, crucially contributes to genome maintenance in meiocytes. Synapsis defects induce oocyte apoptosis ostensibly due to unrepaired DSBs that persist in asynaptic chromosomes. In mice, SPO11-deficient oocytes feature asynapsis, apoptosis and, surprisingly, numerous foci of the ssDNA-binding recombinase RAD51, indicative of DSBs of unknown origin. Hence, asynapsis is suggested to trigger apoptosis due to inefficient DSB repair even in mutants that lack programmed DSBs. By directly detecting ssDNAs, we discovered that RAD51 is an unreliable marker for DSBs in oocytes. Further, SPO11-deficient oocytes have fewer persistent ssDNAs than wild-type oocytes. These observations suggest that oocyte quality is safeguarded in mammals by a synapsis surveillance mechanism that can operate without persistent ssDNAs.
Collapse
Affiliation(s)
- Ramya Ravindranathan
- Institute of Physiological Chemistry, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Kavya Raveendran
- Institute of Physiological Chemistry, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Frantzeskos Papanikos
- Institute of Physiological Chemistry, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Pedro A San-Segundo
- Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Salamanca, Spain
| | - Attila Tóth
- To whom correspondence should be addressed. Tel: +49 351 458 6467; Fax: +49 351 458 6305;
| |
Collapse
|
36
|
Ali H, Unar A, Zubair M, Dil S, Ullah F, Khan I, Hussain A, Shi Q. In silico analysis of a novel pathogenic variant c.7G > A in C14orf39 gene identified by WES in a Pakistani family with azoospermia. Mol Genet Genomics 2022; 297:719-730. [PMID: 35305148 DOI: 10.1007/s00438-022-01876-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/20/2022] [Indexed: 11/25/2022]
Abstract
Infertility is a multifactorial disorder that affects approximately 12% of couples of childbearing ages worldwide. Few studies have been conducted to understand the genetic causes of infertility in depth. The synaptonemal complex (SC), which is essential for the progression of meiosis, is a conserved tripartite structure that binds homologous chromosomes together and is thus required for fertility. This study investigated genetic causes of infertility in a Pakistani consanguineous family containing two patients suffering from non-obstructive azoospermia (NOA). We performed whole-exome sequencing, followed by Sanger sequencing, and identified a novel pathogenic variant (c.7G > A [p.D3N]) in the SC coding gene C14orf39, which was recessively co-segregated with NOA. In silico analysis revealed that charges on wild-type residues were lost, which may result in loss of interactions with other molecules and residues, and a reduction in protein stability occurred, which was caused by the p.D3N mutation. The novel variant generated the mutant protein C14ORF39D3N, and homozygous mutations in C14orf39 resulted in NOA. The transcriptome profile of C14ORF39 shows that it is specifically expressed in early brain development, which suggests that research in this area is required to study other functions of C14ORF39 in addition to its role in the germline. This research highlights the conserved role of C14orf39/SIX6OS1 in assembly of the SC and its indispensable role in facilitating genetic diagnosis in patients with infertility, which may enable the development of future treatments.
Collapse
Affiliation(s)
- Haider Ali
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Ahsanullah Unar
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Muhammad Zubair
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Sobia Dil
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Farman Ullah
- Center of Biotechnology and Microbiology, University of Swat, Swat, 19120, Pakistan
| | - Ihsan Khan
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Ansar Hussain
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China
| | - Qinghua Shi
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
37
|
Nabi S, Askari M, Rezaei-Gazik M, Salehi N, Almadani N, Tahamtani Y, Totonchi M. A rare frameshift mutation in SYCP1 is associated with human male infertility. Mol Hum Reprod 2022; 28:6563198. [PMID: 35377450 DOI: 10.1093/molehr/gaac009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/14/2022] [Indexed: 11/12/2022] Open
Abstract
Proper assembly of the synaptonemal complex is essential for successful meiosis, and impairments in the process lead to infertility. Meiotic transverse filament proteins encoded by the SYCP1 (synaptonemal complex protein 1) gene are one of the main components of the synaptonemal complex and play an important role in correct synapsis and recombination. Family-based whole exome sequencing revealed a rare homozygous SYCP1 frameshift mutation (c.2892delA: p.K967Nfs*1) in two men with severe oligozoospermia, followed by validation and segregation through Sanger sequencing. This single nucleotide deletion not only changes lysine 967 (K) into asparagine (N) but also causes a premature stop codon, which leads to deletion of 968-976 residues from the end of the C-tail region of the SYCP1 protein. Although, sycp1 knockout male mice are reported to be sterile with a complete lack of spermatids and spermatozoa, to date no SYCP1 variant has been associated with human oligozoospermia. HADDOCK analysis indicated that this mutation decreases the ability of the truncated SYCP1 protein to bind DNA. Immunodetection of ϒH2AX signal, in SYCP1 mutant semen cells and a 40% DNA fragmentation index might indicate that a small number of DNA double-strand breaks, which require SYCP1 and/or synapsis to be repaired, are not efficiently repaired, resulting in defects in differentiation of germline cells and appearance of the oligozoospermia phenotype. To our knowledge, this is the first report of homozygous SYCP1 mutation that decreases sperm count. Further studies are required to determine the function of the SYCP1 mutation, which is potentially associated with human oligozoospermia.
Collapse
Affiliation(s)
- Soheila Nabi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Masomeh Askari
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases,Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rezaei-Gazik
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Navid Almadani
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
38
|
Gao W, Yu CX, Zhou WW, Zhang BL, Chambers EA, Dahn HA, Jin JQ, Murphy RW, Zhang YP, Che J. Species persistence with hybridization in toad-headed lizards driven by divergent selection and low recombination. Mol Biol Evol 2022; 39:6561330. [PMID: 35356979 PMCID: PMC9007161 DOI: 10.1093/molbev/msac064] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Speciation plays a central role in evolutionary studies, and particularly how reproductive isolation (RI) evolves. The origins and persistence of RI are distinct processes that require separate evaluations. Treating them separately clarifies the drivers of speciation and then it is possible to link the processes to understand large-scale patterns of diversity. Recent genomic studies have focused predominantly on how species or RI originate. However, we know little about how species persist in face of gene flow. Here, we evaluate a contact zone of two closely related toad-headed lizards (Phrynocephalus) using a chromosome-level genome assembly and population genomics. To some extent, recent asymmetric introgression from Phrynocephalus putjatai to P. vlangalii reduces their genomic differences. However, their highly divergent regions (HDRs) have heterogeneous distributions across the genomes. Functional gene annotation indicates that many genes within HDRs are involved in reproduction and RI. Compared with allopatric populations, contact areas exhibit recent divergent selection on the HDRs and a lower population recombination rate. Taken together, this implies that divergent selection and low genetic recombination help maintain RI. This study provides insights into the genomic mechanisms that drive RI and two species persistence in the face of gene flow during the late stage of speciation.
Collapse
Affiliation(s)
- Wei Gao
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chuan-Xin Yu
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Wei-Wei Zhou
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Bao-Lin Zhang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - E Anne Chambers
- Department of Integrative Biology and Biodiversity Center, University of Texas at Austin, Austin, USA.,Department of Environmental Science, Policy, and Management, Univerity of California, Berkeley, USA
| | - Hollis A Dahn
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jie-Qiong Jin
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Robert W Murphy
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada.,Centre for Biodiversity and Conservation Biology, Royal Ontario Museum, Toronto, Ontario, Canada
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Jing Che
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Key Laboratory of Biodiversity and Ecological Security of Gaoligong Mountain, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
39
|
Xu J, Gao J, Liu J, Huang X, Zhang H, Ma A, Ye J, Zhang X, Li Y, Yang G, Yin H, Khan R, Li T, Fan S, Jiang X, Zhang Y, Jiang H, Ma H, Shi Q. ZFP541 maintains the repression of pre-pachytene transcriptional programs and promotes male meiosis progression. Cell Rep 2022; 38:110540. [PMID: 35320728 DOI: 10.1016/j.celrep.2022.110540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/24/2021] [Accepted: 02/25/2022] [Indexed: 11/28/2022] Open
Abstract
The DSB machinery, which induces the programmed DNA double-strand breaks (DSBs) in the leptotene and zygotene stages during meiosis, is suppressed before the onset of the pachytene stage. However, the biological significance and underlying mechanisms remain largely unclear. Here, we report that ZFP541 is indispensable for the suppression of DSB formation after mid-pachytene. The deletion of Zfp541 in mice causes the aberrant recruitment of DSB machinery to chromosome axes and generation of massive DSBs in late pachytene and diplotene spermatocytes, leading to meiotic arrest at the diplotene stage. Integrated analysis of single-cell RNA sequencing (scRNA-seq) and chromatin immunoprecipitation (ChIP) sequencing data indicate that ZFP541 predominantly binds to promoters of pre-pachytene genes, including meiotic DSB formation-related genes (e.g., Prdm9 and Mei1) and their upstream activators (e.g., Meiosin and Rxra), and maintains their repression in pachytene spermatocytes. Our results reveal that ZFP541 functions as a transcriptional regulator in pachytene spermatocytes, orchestrating the transcriptome to ensure meiosis progression.
Collapse
Affiliation(s)
- Jianze Xu
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Jianing Gao
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Junyan Liu
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Xue Huang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Huan Zhang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Ao Ma
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Jingwei Ye
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Xingxia Zhang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Yang Li
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Gang Yang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Hao Yin
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Ranjha Khan
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Tao Li
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Suixing Fan
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Xiaohua Jiang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Yuanwei Zhang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China
| | - Hanwei Jiang
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China.
| | - Hui Ma
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China.
| | - Qinghua Shi
- Division of Reproduction and Genetics, First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
40
|
Hou D, Yao C, Xu B, Luo W, Ke H, Li Z, Qin Y, Guo T. Variations of C14ORF39 and SYCE1 Identified in Idiopathic Premature Ovarian Insufficiency and Nonobstructive Azoospermia. J Clin Endocrinol Metab 2022; 107:724-734. [PMID: 34718620 DOI: 10.1210/clinem/dgab777] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Indexed: 12/16/2022]
Abstract
CONTEXT Premature ovarian insufficiency (POI) and nonobstructive azoospermia (NOA) are the most severe diseases causing irreversible infertility in females and males, respectively. The contribution of synaptonemal complex (SC) gene variations in the pathogenesis of sporadic patients with POI and NOA has not been systematically illustrated. OBJECTIVE To investigate the role of SC genes in the pathogenesis of sporadic POI and NOA. DESIGN Genetic and functional study. SETTING University-based reproductive medicine center. PATIENT(S) A total of 1030 patients with sporadic POI and 400 patients with sporadic NOA. INTERVENTION(S) The variations of SC genes were filtered in the in-house database of whole exome sequencing performed in 1030 patients with sporadic POI and 400 patients with sporadic NOA. The pathogenic or likely pathogenic variations following recessive inheritance mode were selected according to American College of Medical Genetics and Genomics (ACMG) guidelines and confirmed by Sanger sequencing. The pathogenic effects of the variations were verified by functional studies. MAIN OUTCOME MEASURE(S) ACMG classification and functional characteristics. RESULT(S) Two homozygous variations of C14ORF39 and 2 recessive variations of SYCE1 were first identified in sporadic patients with POI and NOA, respectively. Functional studies showed the C14ORF39 variations significantly accelerated the protein degradation and the variations in SYCE1 disrupted its interaction with SYCP1 or C14ORF39, both of which affected SC assembly and meiosis. CONCLUSION(S) Our study identified novel pathogenic variations of C14ORF39 and SYCE1 in sporadic patients with POI or NOA, highlighting the essential role of SC genes in the maintenance of ovarian and testicular function.
Collapse
Affiliation(s)
- Dong Hou
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Suzhou Research Institute, Shandong University, Suzhou 215123, Jiangsu, China
| | - Chencheng Yao
- Department of Andrology, Center for Men's Health, Shanghai General Hospital; Department of ART, Institute of Urology, Urologic Medical Center, Shanghai General Hospital; Shanghai Key Lab of Reproductive Medicine; Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Bingying Xu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
| | - Wei Luo
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
| | - Hanni Ke
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
| | - Zheng Li
- Department of Andrology, Center for Men's Health, Shanghai General Hospital; Department of ART, Institute of Urology, Urologic Medical Center, Shanghai General Hospital; Shanghai Key Lab of Reproductive Medicine; Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
| | - Ting Guo
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan 250012, Shandong, China
- Reproductive Hospital Affiliated to Shandong University, Jinan 250001, Shandong, China
| |
Collapse
|
41
|
Pyatnitskaya A, Andreani J, Guérois R, De Muyt A, Borde V. The Zip4 protein directly couples meiotic crossover formation to synaptonemal complex assembly. Genes Dev 2022; 36:53-69. [PMID: 34969823 PMCID: PMC8763056 DOI: 10.1101/gad.348973.121] [Citation(s) in RCA: 21] [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] [Received: 08/27/2021] [Accepted: 12/08/2021] [Indexed: 11/24/2022]
Abstract
Meiotic recombination is triggered by programmed double-strand breaks (DSBs), a subset of these being repaired as crossovers, promoted by eight evolutionarily conserved proteins, named ZMM. Crossover formation is functionally linked to synaptonemal complex (SC) assembly between homologous chromosomes, but the underlying mechanism is unknown. Here we show that Ecm11, a SC central element protein, localizes on both DSB sites and sites that attach chromatin loops to the chromosome axis, which are the starting points of SC formation, in a way that strictly requires the ZMM protein Zip4. Furthermore, Zip4 directly interacts with Ecm11, and point mutants that specifically abolish this interaction lose Ecm11 binding to chromosomes and exhibit defective SC assembly. This can be partially rescued by artificially tethering interaction-defective Ecm11 to Zip4. Mechanistically, this direct connection ensuring SC assembly from CO sites could be a way for the meiotic cell to shut down further DSB formation once enough recombination sites have been selected for crossovers, thereby preventing excess crossovers. Finally, the mammalian ortholog of Zip4, TEX11, also interacts with the SC central element TEX12, suggesting a general mechanism.
Collapse
Affiliation(s)
- Alexandra Pyatnitskaya
- Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, Dynamics of Genetic Information, UMR3244, Centre National de la Recherche Scientifique (CNRS), Paris 75248, France
| | - Jessica Andreani
- Université Paris-Saclay, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette 91198, France
| | - Raphaël Guérois
- Université Paris-Saclay, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette 91198, France
| | - Arnaud De Muyt
- Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, Dynamics of Genetic Information, UMR3244, Centre National de la Recherche Scientifique (CNRS), Paris 75248, France
| | - Valérie Borde
- Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, Dynamics of Genetic Information, UMR3244, Centre National de la Recherche Scientifique (CNRS), Paris 75248, France
| |
Collapse
|
42
|
Xie C, Wang W, Tu C, Meng L, Lu G, Lin G, Lu LY, Tan YQ. OUP accepted manuscript. Hum Reprod Update 2022; 28:763-797. [PMID: 35613017 DOI: 10.1093/humupd/dmac024] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/18/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Chunbo Xie
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Weili Wang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Chaofeng Tu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lanlan Meng
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Guangxiu Lu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ge Lin
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lin-Yu Lu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue-Qiu Tan
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
43
|
Ishiguro KI, Shimada R. MEIOSIN directs initiation of meiosis and subsequent meiotic prophase program during spermatogenesis. Genes Genet Syst 2021; 97:27-39. [PMID: 34955498 DOI: 10.1266/ggs.21-00054] [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: 11/23/2022] Open
Abstract
Meiosis is a crucial process for spermatogenesis and oogenesis. Initiation of meiosis coincides with spermatocyte differentiation and is followed by meiotic prophase, a prolonged G2 phase that ensures the completion of numerous meiosis-specific chromosome events. During meiotic prophase, chromosomes are organized into axis-loop structures, which underlie meiosis-specific events such as meiotic recombination and homolog synapsis. In spermatocytes, meiotic prophase is accompanied by robust alterations of gene expression programs and chromatin status for subsequent sperm production. The mechanisms regulating meiotic initiation and subsequent meiotic prophase programs are enigmatic. Recently, we discovered MEIOSIN (Meiosis initiator), a DNA-binding protein that directs the switch from mitosis to meiosis. This review mainly focuses on how MEIOSIN is involved in meiotic initiation and the meiotic prophase program during spermatogenesis. Further, we discuss the downstream genes activated by MEIOSIN, which are crucial for meiotic prophase-specific events, from the viewpoint of chromosome dynamics and the gene expression program.
Collapse
Affiliation(s)
- Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University
| | - Ryuki Shimada
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University
| |
Collapse
|
44
|
Li W, Zhang L, Shinohara A, Keeney S. Editorial: Meiosis: From Molecular Basis to Medicine. Front Cell Dev Biol 2021; 9:812292. [PMID: 34926477 PMCID: PMC8671932 DOI: 10.3389/fcell.2021.812292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Wei Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Liangran Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Akira Shinohara
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, Howard Hughes Medical Institute, New York, NY, United States
| |
Collapse
|
45
|
Qu W, Liu C, Xu YT, Xu YM, Luo MC. The formation and repair of DNA double-strand breaks in mammalian meiosis. Asian J Androl 2021; 23:572-579. [PMID: 34708719 PMCID: PMC8577251 DOI: 10.4103/aja202191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Programmed DNA double-strand breaks (DSBs) are necessary for meiosis in mammals. A sufficient number of DSBs ensure the normal pairing/synapsis of homologous chromosomes. Abnormal DSB repair undermines meiosis, leading to sterility in mammals. The DSBs that initiate recombination are repaired as crossovers and noncrossovers, and crossovers are required for correct chromosome separation. Thus, the placement, timing, and frequency of crossover formation must be tightly controlled. Importantly, mutations in many genes related to the formation and repair of DSB result in infertility in humans. These mutations cause nonobstructive azoospermia in men, premature ovarian insufficiency and ovarian dysgenesis in women. Here, we have illustrated the formation and repair of DSB in mammals, summarized major factors influencing the formation of DSB and the theories of crossover regulation.
Collapse
Affiliation(s)
- Wei Qu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Cong Liu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Ya-Ting Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yu-Min Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Meng-Cheng Luo
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
46
|
Imai Y, Olaya I, Sakai N, Burgess SM. Meiotic Chromosome Dynamics in Zebrafish. Front Cell Dev Biol 2021; 9:757445. [PMID: 34692709 PMCID: PMC8531508 DOI: 10.3389/fcell.2021.757445] [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] [Received: 08/12/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Recent studies in zebrafish have revealed key features of meiotic chromosome dynamics, including clustering of telomeres in the bouquet configuration, biogenesis of chromosome axis structures, and the assembly and disassembly of the synaptonemal complex that aligns homologs end-to-end. The telomere bouquet stage is especially pronounced in zebrafish meiosis and sub-telomeric regions play key roles in mediating pairing and homologous recombination. In this review, we discuss the temporal progression of these events in meiosis prophase I and highlight the roles of proteins associated with meiotic chromosome architecture in homologous recombination. Finally, we discuss the interplay between meiotic mutants and gonadal sex differentiation and future research directions to study meiosis in living cells, including cell culture.
Collapse
Affiliation(s)
- Yukiko Imai
- Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
| | - Ivan Olaya
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, United States.,Integrative Genetics and Genomics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Noriyoshi Sakai
- Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan.,Department of Genetics, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Japan
| | - Sean M Burgess
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
47
|
Abstract
The specialized two-stage meiotic cell division program halves a cell's chromosome complement in preparation for sexual reproduction. This reduction in ploidy requires that in meiotic prophase, each pair of homologous chromosomes (homologs) identify one another and form physical links through DNA recombination. Here, we review recent advances in understanding the complex morphological changes that chromosomes undergo during meiotic prophase to promote homolog identification and crossing over. We focus on the structural maintenance of chromosomes (SMC) family cohesin complexes and the meiotic chromosome axis, which together organize chromosomes and promote recombination. We then discuss the architecture and dynamics of the conserved synaptonemal complex (SC), which assembles between homologs and mediates local and global feedback to ensure high fidelity in meiotic recombination. Finally, we discuss exciting new advances, including mechanisms for boosting recombination on particular chromosomes or chromosomal domains and the implications of a new liquid crystal model for SC assembly and structure. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sarah N Ur
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093, USA; ,
| | - Kevin D Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093, USA; , .,Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
48
|
The synaptonemal complex imposes crossover interference and heterochiasmy in Arabidopsis. Proc Natl Acad Sci U S A 2021; 118:2023613118. [PMID: 33723072 PMCID: PMC8000504 DOI: 10.1073/pnas.2023613118] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Meiotic recombination promotes genetic diversity by shuffling parental chromosomes. As observed by the very first geneticists, crossovers inhibit the formation of another crossover nearby, an elusive phenomenon called crossover interference. Another intriguing observation is heterochiasmy, the marked difference in male and female crossover rates observed in many species. Here, we show that the synaptonemal complex, a structure that zips homologous chromosomes together during meiosis, is essential for crossover interference in Arabidopsis. This suggests that a signal that inhibits crossover formation nearby a first crossover propagates along this specific structure. Furthermore, in the absence of the synaptonemal complex, crossover frequencies become identical in both sexes, suggesting that heterochiasmy is due to variation of crossover interference imposed by the synaptonemal complex. Meiotic crossovers (COs) have intriguing patterning properties, including CO interference, the tendency of COs to be well-spaced along chromosomes, and heterochiasmy, the marked difference in male and female CO rates. During meiosis, transverse filaments transiently associate the axes of homologous chromosomes, a process called synapsis that is essential for CO formation in many eukaryotes. Here, we describe the spatial organization of the transverse filaments in Arabidopsis (ZYP1) and show it to be evolutionary conserved. We show that in the absence of ZYP1 (zyp1a zyp1b null mutants), chromosomes associate in pairs but do not synapse. Unexpectedly, in absence of ZYP1, CO formation is not prevented but increased. Furthermore, genome-wide analysis of recombination revealed that CO interference is abolished, with the frequent observation of close COs. In addition, heterochiasmy was erased, with identical CO rates in males and females. This shows that the tripartite synaptonemal complex is dispensable for CO formation and has a key role in regulating their number and distribution, imposing CO interference and heterochiasmy.
Collapse
|
49
|
Cooke PS, Walker WH. Male fertility in mice requires classical and nonclassical androgen signaling. Cell Rep 2021; 36:109557. [PMID: 34407397 DOI: 10.1016/j.celrep.2021.109557] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/03/2021] [Accepted: 07/27/2021] [Indexed: 11/22/2022] Open
Abstract
Molecular mechanisms by which androgens signal through the androgen receptor (AR) to maintain male fertility are poorly understood. Transgenic mice were produced expressing mutant ARs that can only (1) alter gene transcription through the classical response pathway (AR-C) or (2) activate kinase signaling cascades via the nonclassical pathway (AR-NC). AR-C is sufficient to produce sperm and fertility. Haploid germ cell production, the blood-testis barrier, and spermatid migration are supported by AR-NC. Gene expression essential for chromosome synapsis during meiosis requires AR-C. We identify targets of androgen signaling required for male fertility and provide a mechanistic explanation for meiotic germ cell arrest in the absence of androgen signaling. Prostate differentiation occurs with AR-C alone, but full development requires synergistic nonclassical signaling. Both AR signaling pathways are necessary for normal male reproductive tract development and function, validating our mouse models for studies of AR functions in other target tissues.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - William H Walker
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Magee-Womens Research Institute, Pittsburgh, PA 15213, USA.
| |
Collapse
|
50
|
Structural basis of meiotic chromosome synaptic elongation through hierarchical fibrous assembly of SYCE2-TEX12. Nat Struct Mol Biol 2021; 28:681-693. [PMID: 34373646 PMCID: PMC7612376 DOI: 10.1038/s41594-021-00636-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/30/2021] [Indexed: 01/16/2023]
Abstract
The synaptonemal complex (SC) is a supramolecular protein assembly that mediates synapsis between homologous chromosomes during meiosis. SC elongation along the chromosome length (up to 24 μm) depends on its midline α-fibrous component SYCE2-TEX12. Here, we report X-ray crystal structures of human SYCE2-TEX12 as an individual building-block and upon assembly within a fibrous lattice. We combine these structures with mutagenesis, biophysics and electron microscopy to reveal the hierarchical mechanism of SYCE2-TEX12 fibre assembly. SYCE2-TEX12’s building-blocks are 2:2 coiled-coils which dimerise into 4:4 hetero-oligomers and interact end-to-end and laterally to form 10-nm fibres, which intertwine within 40-nm bundled micrometre-long fibres that define the SC’s midline structure. This assembly mechanism bears striking resemblance with intermediate filament proteins vimentin, lamin and keratin. Thus, SYCE2-TEX12 exhibits behaviour typical of cytoskeletal proteins to provide an α-fibrous SC backbone that structurally underpins synaptic elongation along meiotic chromosomes.
Collapse
|