1
|
Arter M, Keeney S. Divergence and conservation of the meiotic recombination machinery. Nat Rev Genet 2024; 25:309-325. [PMID: 38036793 DOI: 10.1038/s41576-023-00669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 12/02/2023]
Abstract
Sexually reproducing eukaryotes use recombination between homologous chromosomes to promote chromosome segregation during meiosis. Meiotic recombination is almost universally conserved in its broad strokes, but specific molecular details often differ considerably between taxa, and the proteins that constitute the recombination machinery show substantial sequence variability. The extent of this variation is becoming increasingly clear because of recent increases in genomic resources and advances in protein structure prediction. We discuss the tension between functional conservation and rapid evolutionary change with a focus on the proteins that are required for the formation and repair of meiotic DNA double-strand breaks. We highlight phylogenetic relationships on different time scales and propose that this remarkable evolutionary plasticity is a fundamental property of meiotic recombination that shapes our understanding of molecular mechanisms in reproductive biology.
Collapse
Affiliation(s)
- Meret Arter
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Santiago J, Silva JV, Santos MAS, Fardilha M. Age-Dependent Alterations in Semen Parameters and Human Sperm MicroRNA Profile. Biomedicines 2023; 11:2923. [PMID: 38001924 PMCID: PMC10669352 DOI: 10.3390/biomedicines11112923] [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: 10/16/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The trend to delay parenthood is increasing, impacting fertility and reproductive outcomes. Advanced paternal age (APA), defined as men's age above 40 years at conception, has been linked with testicular impairment, abnormal semen parameters, and poor reproductive and birth outcomes. Recently, the significance of sperm microRNA for fertilization and embryonic development has emerged. This work aimed to investigate the effects of men's age on semen parameters and sperm microRNA profiles. The ejaculates of 333 Portuguese men were collected between 2018 and 2022, analyzed according to WHO guidelines, and a density gradient sperm selection was performed. For microRNA expression analysis, 16 normozoospermic human sperm samples were selected and divided into four age groups: ≤30, 31-35, 36-40, and >40 years. microRNA target genes were retrieved from the miRDB and TargetScan databases and Gene Ontology analysis was performed using the DAVID tool. No significant correlation was found between male age and conventional semen parameters, except for volume. Fifteen differentially expressed microRNAs (DEMs) between groups were identified. Enrichment analysis suggested the involvement of DEMs in the sperm of men with advanced age in critical biological processes like embryonic development, morphogenesis, and male gonad development. Targets of DEMs were involved in signaling pathways previously associated with the ageing process, including cellular senescence, autophagy, insulin, and mTOR pathways. These results suggest that although conventional semen parameters were not affected by men's age, alterations in microRNA regulation may occur and be responsible for poor fertility and reproductive outcomes associated with APA.
Collapse
Affiliation(s)
- Joana Santiago
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| | - Joana V. Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| | - Manuel A. S. Santos
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
- Multidisciplinary Institute of Ageing, MIA-Portugal, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| |
Collapse
|
3
|
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
|
4
|
Xiong J, Bao J, Hu W, Shang M, Zhang L. Whole-genome resequencing reveals genetic diversity and selection characteristics of dairy goat. Front Genet 2023; 13:1044017. [PMID: 36685859 PMCID: PMC9852865 DOI: 10.3389/fgene.2022.1044017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
The dairy goat is one of the earliest dairy livestock species, which plays an important role in the economic development, especially for developing countries. With the development of agricultural civilization, dairy goats have been widely distributed across the world. However, few studies have been conducted on the specific characteristics of dairy goat. In this study, we collected the whole-genome data of 89 goat individuals by sequencing 48 goats and employing 41 publicly available goats, including five dairy goat breeds (Saanen, Nubian, Alpine, Toggenburg, and Guanzhong dairy goat; n = 24, 15, 11, 6, 6), and three goat breeds (Guishan goat, Longlin goat, Yunshang Black goat; n = 6, 15, 6). Through compared the genomes of dairy goat and non-dairy goat to analyze genetic diversity and selection characteristics of dairy goat. The results show that the eight goats could be divided into three subgroups of European, African, and Chinese indigenous goat populations, and we also found that Australian Nubian, Toggenburg, and Australian Alpine had the highest linkage disequilibrium, the lowest level of nucleotide diversity, and a higher inbreeding coefficient, indicating that they were strongly artificially selected. In addition, we identified several candidate genes related to the specificity of dairy goat, particularly genes associated with milk production traits (GHR, DGAT2, ELF5, GLYCAM1, ACSBG2, ACSS2), reproduction traits (TSHR, TSHB, PTGS2, ESR2), immunity traits (JAK1, POU2F2, LRRC66). Our results provide not only insights into the evolutionary history and breed characteristics of dairy goat, but also valuable information for the implementation and improvement of dairy goat cross breeding program.
Collapse
|
5
|
Xu Y, Chen Z, Wu P, Qu W, Shi H, Cheng M, Xu Y, Jin T, Liu C, Liu C, Li Y, Luo M. Nuclear localization of human MEIOB requires its NLS in the OB domain and interaction with SPATA22. Acta Biochim Biophys Sin (Shanghai) 2022; 55:154-161. [PMID: 36331299 PMCID: PMC10157540 DOI: 10.3724/abbs.2022156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MEIOB is a vital protein in meiotic homologous recombination and plays an indispensable role in human gametogenesis. In mammals, MEIOB and its partner SPATA22 form a heterodimer, ensuring their effective localization on single-strand DNA (ssDNA) and proper synapsis processes. Mutations in human MEIOB (hMEIOB) cause human infertility attributed to the failure of its interaction with human SPATA22 (hSPATA22) and ssDNA binding. However, the detailed mechanism is still unclear. In our study, truncated or full-length hMEIOB and hSPATA22 are traced by fused expression with fluorescent proteins (i.e., copGFP or mCherry), and the live cell imaging system is used to observe the expression and localization of the proteins. When transfected alone, hMEIOB accumulates in the cytoplasm. Interestingly, a covered NLS in the OB domain of hMEIOB is identified, which can be exposed by hSPATA22 and is necessary for the nuclear localization of hMEIOB. When hSPATA22 loses its hMEIOB interacting domain or NLS, the nuclear localization of hMEIOB is aborted. Collectively, our results prove that the NLS in the OB domain of hMEIOB and interaction with hSPATA22 are required for hMEIOB nuclear localization.
Collapse
|
6
|
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: 5.0] [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
|
7
|
Gao D, Huang J, Lin G, Lu J. A time-course transcriptome analysis of gonads from yellow catfish (Pelteobagrus fulvidraco) reveals genes associated with gonad development. BMC Genomics 2022; 23:409. [PMID: 35637435 PMCID: PMC9153201 DOI: 10.1186/s12864-022-08651-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background The yellow catfish, Pelteobagrus fulvidraco, is a commercially important fish species. It is widely distributed in the fresh water areas of China, including rivers, lakes, and reservoirs. Like many other aquaculture fish species, people have observed significant size dimorphism between male and female yellow catfish and it shows a growth advantage in males. Results Here, at the first time, the time-course transcriptome was used to explore the various expression profiles of genes in different gonad developmental stages and genders. A total of 2696 different expression genes (DEGs) were identified from different stages. Based on these DEGs, 13 gonad development related genes were identified which showed time-specific or sex biased expression patterns. Conclusion This study will provide the crucial information on the molecular mechanism of gonad development of female and male yellow catfish. Especially, during the different gonad development stages, these 13 gonad development related genes exhibit various expression patterns in female and male individual respectively. These results could inspire and facilitate us to understanding the various roles of these genes play in different gonad development stages and genders. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08651-0.
Collapse
Affiliation(s)
- Dong Gao
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082, China
| | - Junrou Huang
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082, China
| | - Genmei Lin
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082, China
| | - Jianguo Lu
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082, China. .,Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519080, China. .,Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, 510275, Guangdong, China. .,Pearl River Estuary Marine Ecosystem Research Station, Ministry of Education, Zhuhai, 519000, China.
| |
Collapse
|
8
|
Yao C, Hou D, Ji Z, Pang D, Li P, Tian R, Zhang Y, Ou N, Bai H, Zhi E, Huang Y, Qin Y, Zhao J, Wang C, Zhou Z, Guo T, Li Z. Bi‐allelic
SPATA22
Variants Cause Premature Ovarian Insufficiency and Non‐obstructive Azoospermia Due to Meiotic Arrest. Clin Genet 2022; 101:507-516. [DOI: 10.1111/cge.14129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Chencheng Yao
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- School of Life Science and Technology ShanghaiTech University Shanghai China
| | - Dong Hou
- Center for Reproductive Medicine Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key laboratory of Reproductive Endocrinology of Ministry of Education, and Shandong Provincial Clinical Medicine Research Center for Reproductive Health Jinan China
| | - Zhiyong Ji
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- School of Life Science and Technology ShanghaiTech University Shanghai China
- State Key Lab of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Dongmei Pang
- Jimo Hospital of Traditional Chinese Medicine Affiliated to Shandong University of Traditional Chinese Medicine Qingdao China
| | - Peng Li
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ruhui Tian
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yuxiang Zhang
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ningjing Ou
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- School of Life Science and Technology ShanghaiTech University Shanghai China
- State Key Lab of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Haowei Bai
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Erlei Zhi
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yuhua Huang
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yingying Qin
- Center for Reproductive Medicine Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key laboratory of Reproductive Endocrinology of Ministry of Education, and Shandong Provincial Clinical Medicine Research Center for Reproductive Health Jinan China
| | - Jingpeng Zhao
- State Key Lab of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Chenchen Wang
- Shanghai Advanced Research Institute, Stem Cell and Reproductive Biology Laboratory Chinese Academy of Sciences Shanghai China
| | - Zhi Zhou
- School of Life Science and Technology ShanghaiTech University Shanghai China
| | - Ting Guo
- Center for Reproductive Medicine Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key laboratory of Reproductive Endocrinology of Ministry of Education, and Shandong Provincial Clinical Medicine Research Center for Reproductive Health Jinan China
| | - Zheng Li
- Department of Andrology, Center for Men's Health, Department of ART, Institute of Urology, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- State Key Lab of Reproductive Medicine Nanjing Medical University Nanjing China
| |
Collapse
|
9
|
Ishiguro KI. Sexually Dimorphic Properties in Meiotic Chromosome. Sex Dev 2022; 16:423-434. [PMID: 35130542 DOI: 10.1159/000520682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 10/22/2021] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Meiosis is a crucial process for germ cell development. It consists of 1 round of DNA replication followed by 2 rounds of chromosome segregation, producing haploid gametes from diploid cells. During meiotic prophase, chromosomes are organized into axis-loop structures, which underlie meiosis-specific events such as meiotic recombination and homolog synapsis. Meiosis-specific cohesin plays a pivotal role in establishing higher-order chromosome architecture and regulating chromosome dynamics. SUMMARY Notably, sexually dimorphic properties of chromosome architecture are prominent during meiotic prophase, despite the same axial proteins being conserved between male and female. The difference in chromosome structure between the sexes gives sexual differences in the regulation of meiotic recombination and crossover distribution. KEY MESSAGES This review mainly focuses on the sexual differences of meiosis from the viewpoint of chromosome structure in mammals, elucidating the differences in meiotic recombination and homolog synapsis between the sexes.
Collapse
Affiliation(s)
- Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
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: 28] [Impact Index Per Article: 14.0] [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
|
11
|
Ghieh F, Barbotin AL, Swierkowski-Blanchard N, Leroy C, Fortemps J, Gerault C, Hue C, Mambu Mambueni H, Jaillard S, Albert M, Bailly M, Izard V, Molina-Gomes D, Marcelli F, Prasivoravong J, Serazin V, Dieudonne MN, Delcroix M, Garchon HJ, Louboutin A, Mandon-Pepin B, Ferlicot S, Vialard F. OUP accepted manuscript. Hum Reprod 2022; 37:1334-1350. [PMID: 35413094 PMCID: PMC9156845 DOI: 10.1093/humrep/deac057] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/07/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- F Ghieh
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - A L Barbotin
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, Hôpital Jeanne de Flandre, Centre Hospitalier et Universitaire, Lille, France
| | - N Swierkowski-Blanchard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- Département de Gynécologie Obstétrique, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - C Leroy
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, Hôpital Jeanne de Flandre, Centre Hospitalier et Universitaire, Lille, France
| | - J Fortemps
- Service d’Anatomie Pathologique, CHI de Poissy/Saint-Germain-en-Laye, Saint-Germain-en-Laye, France
| | - C Gerault
- Département de Génétique, Laboratoire de Biologie Médicale, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - C Hue
- Department of Biotechnology and Health, UVSQ, Université Paris-Saclay, Inserm UMR 1173, Montigny-le-Bretonneux, France
| | - H Mambu Mambueni
- Department of Biotechnology and Health, UVSQ, Université Paris-Saclay, Inserm UMR 1173, Montigny-le-Bretonneux, France
| | - S Jaillard
- Service de Cytogénétique, CHU Rennes, Rennes, France
- INSERM, EHESP, IRSET—UMR_S 1085, Université Rennes 1, Rennes, France
| | - M Albert
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - M Bailly
- Département de Gynécologie Obstétrique, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - V Izard
- Service d’Urologie, AP-HP, Université Paris-Saclay, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - D Molina-Gomes
- Département de Génétique, Laboratoire de Biologie Médicale, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - F Marcelli
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, Hôpital Jeanne de Flandre, Centre Hospitalier et Universitaire, Lille, France
| | - J Prasivoravong
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, Hôpital Jeanne de Flandre, Centre Hospitalier et Universitaire, Lille, France
| | - V Serazin
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- Département de Génétique, Laboratoire de Biologie Médicale, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - M N Dieudonne
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - M Delcroix
- Département de Génétique, Laboratoire de Biologie Médicale, CHI de Poissy/Saint-Germain-en-Laye, Poissy, France
| | - H J Garchon
- Department of Biotechnology and Health, UVSQ, Université Paris-Saclay, Inserm UMR 1173, Montigny-le-Bretonneux, France
| | - A Louboutin
- Service d’Anatomie Pathologique, CHI de Poissy/Saint-Germain-en-Laye, Saint-Germain-en-Laye, France
| | - B Mandon-Pepin
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- École Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - S Ferlicot
- Service d’Anatomie Pathologique, AP-HP, Université Paris-Saclay, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - F Vialard
- Correspondence address. Tel: +33-139-274-700; E-mail:
| |
Collapse
|
12
|
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: 1.7] [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
|
13
|
Wu Y, Li Y, Murtaza G, Zhou J, Jiao Y, Gong C, Hu C, Han Q, Zhang H, Zhang Y, Shi B, Ma H, Jiang X, Shi Q. Whole-exome sequencing of consanguineous families with infertile men and women identifies homologous mutations in SPATA22 and MEIOB. Hum Reprod 2021; 36:2793-2804. [PMID: 34392356 DOI: 10.1093/humrep/deab185] [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: 04/13/2021] [Revised: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Can whole-exome sequencing (WES) reveal pathogenic mutations in two consanguineous Pakistani families with infertile patients? SUMMARY ANSWER A homozygous spermatogenesis associated 22 (SPATA22) frameshift mutation (c.203del), which disrupts the interaction with meiosis specific with OB-fold (MEIOB), and a MEIOB splicing mutation (c.683-1G>A) that led to loss of MEIOB protein cause familial infertility. WHAT IS KNOWN ALREADY MEIOB and SPATA22, direct binding partners and functional collaborators, form a meiosis-specific heterodimer that regulates meiotic recombination. The protein stability and the axial localization of MEIOB and SPATA22 depend on each other. Meiob and Spata22 knockout mice have the same phenotypes: mutant spermatocytes can initiate meiotic recombination but are unable to complete DSB repair, leading to crossover formation failure, meiotic prophase arrest, and sterility. STUDY DESIGN, SIZE, DURATION We performed WES for the patients and controls in two consanguineous Pakistani families to screen for mutations. The pathogenicity of the identified mutations was assessed by in vitro assay and mutant mouse model. PARTICIPANTS/MATERIALS, SETTING, METHODS Two consanguineous Pakistani families with four patients (three men and one woman) suffering from primary infertility were recruited. SPATA22 and MEIOB mutations were screened from the WES data, followed by functional verification in cultured cells and mice. MAIN RESULTS AND THE ROLE OF CHANCE A homozygous SPATA22 frameshift mutation (c.203del) was identified in a patient with non-obstructive azoospermia (NOA) from a consanguineous Pakistani family and a homozygous MEIOB splicing mutation (c.683-1G>A) was identified in two patients with NOA and one infertile woman from another consanguineous Pakistani family. The SPATA22 mutation destroyed the interaction with MEIOB. The MEIOB splicing mutation induced Exon 9 skipping, which causes a 32aa deletion in the oligonucleotide-binding domain without affecting the interaction between MEIOB and SPATA22. Furthermore, analyses of the Meiob mutant mice modelling the patients' mutation revealed that the MEIOB splicing mutation leads to loss of MEIOB proteins, abolished SPATA22 recruitment on chromosome axes, and meiotic arrest due to meiotic recombination failure. Thus, our study suggests that SPATA22 and MEIOB may both be causative genes for human infertility. LIMITATIONS, REASONS FOR CAUTION As SPATA22 and MEIOB are interdependent and essential for meiotic recombination, screening for mutations of SPATA22 and MEIOB in both infertile men and women in larger cohorts is important to further reveal the role of the SPATA22 and MEIOB heterodimer in human fertility. WIDER IMPLICATIONS OF THE FINDINGS These findings provide direct clinical and functional evidence that mutations in SPATA22 and MEIOB can cause meiotic recombination failure, supporting a role for these mutations in human infertility and their potential use as targets for genetic diagnosis of human infertility. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Key Research and Developmental Program of China (2018YFC1003900, 2018YFC1003700, and 2019YFA0802600), the National Natural Science Foundation of China (31890780, 31630050, 32061143006, 82071709, and 31871514), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB19000000). The authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yufan Wu
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Yang Li
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Ghulam Murtaza
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Jianteng Zhou
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Yuying Jiao
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Chenjia Gong
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Congyuan Hu
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Qiqi Han
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Huan Zhang
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Yuanwei Zhang
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Baolu Shi
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Hui Ma
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Xiaohua Jiang
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| | - Qinghua Shi
- First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China
| |
Collapse
|
14
|
Robert N, Yan C, Si-Jiu Y, Bo L, He H, Pengfei Z, Hongwei X, Jian Z, Shijie L, Qian Z. Expression of Rad51 and the histo-morphological evaluation of testis of the sterile male cattle-yak. Theriogenology 2021; 172:239-254. [PMID: 34298284 DOI: 10.1016/j.theriogenology.2021.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/18/2022]
Abstract
Meiotic recombination is key to the repair of DNA double-strand break damage, provide a link between homologs for proper chromosome segregation as well as ensure genetic diversity in organisms. Defects in recombination often lead to sterility. The ubiquitously expressed Rad51 and the meiosis-specific DMC1 are two closely related recombinases that catalyze the key strand invasion and exchange step of meiotic recombination. This study cloned and sequenced the coding region of cattle-yak Rad51 and determined its mRNA and protein expression levels, evaluated its molecular and evolutionary relationship as well as evaluated the histo-morphological structure of testes in the yellow cattle, yak and the sterile cattle-yak hybrid. The Rad51 gene was amplified using PCR, cloned and sequenced using testicular cDNA from yak and cattle-yak. Real-time PCR was used to examine the expression levels of Rad51/DMC1 mRNA in the cattle, yak and cattle-yak testis while western blotting, immunofluorescence and immunohistochemistry were used to assess the protein expression and localization of Rad51/DMC1 protein in the testicular tissue sections. The results revealed that the mRNA and protein expression of Rad51 and DMC1 are extremely low in the male cattle-yak testis with a corresponding higher incidence of germ cell apoptosis. There was also thinning of the germinal epithelium possibly due to the depletion of the germ cells leading to the widening of the lumen area of the cattle-yak seminiferous tubule. Our findings provide support for the hypothesis that the low expression of Rad51 and DMC1 may contribute to the male hybrid sterility in the cattle-yak.
Collapse
Affiliation(s)
- Niayale Robert
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Cui Yan
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine Gansu Agricultural University, Lanzhou, China.
| | - Yu Si-Jiu
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine Gansu Agricultural University, Lanzhou, China
| | - Liao Bo
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine Gansu Agricultural University, Lanzhou, China
| | - Zhao Pengfei
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xu Hongwei
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Zhang Jian
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Li Shijie
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Zhang Qian
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
15
|
Mhaskar AN, Koornneef L, Zelensky AN, Houtsmuller AB, Baarends WM. High Resolution View on the Regulation of Recombinase Accumulation in Mammalian Meiosis. Front Cell Dev Biol 2021; 9:672191. [PMID: 34109178 PMCID: PMC8181746 DOI: 10.3389/fcell.2021.672191] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
A distinguishing feature of meiotic DNA double-strand breaks (DSBs), compared to DSBs in somatic cells, is the fact that they are induced in a programmed and specifically orchestrated manner, which includes chromatin remodeling prior to DSB induction. In addition, the meiotic homologous recombination (HR) repair process that follows, is different from HR repair of accidental DSBs in somatic cells. For instance, meiotic HR involves preferred use of the homolog instead of the sister chromatid as a repair template and subsequent formation of crossovers and non-crossovers in a tightly regulated manner. An important outcome of this distinct repair pathway is the pairing of homologous chromosomes. Central to the initial steps in homology recognition during meiotic HR is the cooperation between the strand exchange proteins (recombinases) RAD51 and its meiosis-specific paralog DMC1. Despite our understanding of their enzymatic activity, details on the regulation of their assembly and subsequent molecular organization at meiotic DSBs in mammals have remained largely enigmatic. In this review, we summarize recent mouse data on recombinase regulation via meiosis-specific factors. Also, we reflect on bulk “omics” studies of initial meiotic DSB processing, compare these with studies using super-resolution microscopy in single cells, at single DSB sites, and explore the implications of these findings for our understanding of the molecular mechanisms underlying meiotic HR regulation.
Collapse
Affiliation(s)
- Aditya N Mhaskar
- Department of Developmental Biology, Erasmus MC, Rotterdam, Netherlands
| | - Lieke Koornneef
- Department of Developmental Biology, Erasmus MC, Rotterdam, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Alex N Zelensky
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands
| | - Adriaan B Houtsmuller
- Erasmus Optical Imaging Centre, Department of Pathology, Erasmus MC, Rotterdam, Netherlands.,Department of Pathology, Erasmus MC, Rotterdam, Netherlands
| | - Willy M Baarends
- Department of Developmental Biology, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
16
|
Takemoto K, Tani N, Takada-Horisawa Y, Fujimura S, Tanno N, Yamane M, Okamura K, Sugimoto M, Araki K, Ishiguro KI. Meiosis-Specific C19orf57/4930432K21Rik/BRME1 Modulates Localization of RAD51 and DMC1 to DSBs in Mouse Meiotic Recombination. Cell Rep 2021; 31:107686. [PMID: 32460033 DOI: 10.1016/j.celrep.2020.107686] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/16/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022] Open
Abstract
Meiotic recombination is critical for genetic exchange and generation of chiasmata that ensures faithful chromosome segregation during meiosis I. Meiotic recombination is initiated by DNA double-strand break (DSB) followed by multiple processes of DNA repair. The exact mechanisms for how recombinases localize to DSB remain elusive. Here, we show that C19orf57/4930432K21Rik/BRME1 is a player for meiotic recombination in mice. C19orf57/4930432K21Rik/BRME1 associates with single-stranded DNA (ssDNA) binding proteins, BRCA2 and MEILB2/HSF2BP, which are critical recruiters of recombinases onto DSB sites. Disruption of C19orf57/4930432K21Rik/BRME1 shows severe impact on DSB repair and male fertility. Remarkably, removal of ssDNA binding proteins from DSB sites is delayed, and reciprocally, the loading of RAD51 and DMC1 onto resected ssDNA is impaired in Brme1 knockout (KO) spermatocytes. We propose that C19orf57/4930432K21Rik/BRME1 modulates localization of recombinases to meiotic DSB sites through the interaction with the BRCA2-MEILB2/HSF2BP complex during meiotic recombination.
Collapse
Affiliation(s)
- Kazumasa Takemoto
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan; Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Naoki Tani
- Liaison Laboratory Research Promotion Center, IMEG, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yuki Takada-Horisawa
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan
| | - Sayoko Fujimura
- Liaison Laboratory Research Promotion Center, IMEG, Kumamoto University, Kumamoto 860-0811, Japan
| | - Nobuhiro Tanno
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan
| | - Mariko Yamane
- RIKEN, Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Kaho Okamura
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan
| | - Michihiko Sugimoto
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kimi Araki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan; Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
17
|
Zhang X, Gunewardena S, Wang N. Nutrient restriction synergizes with retinoic acid to induce mammalian meiotic initiation in vitro. Nat Commun 2021; 12:1758. [PMID: 33741948 PMCID: PMC7979727 DOI: 10.1038/s41467-021-22021-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 02/23/2021] [Indexed: 02/08/2023] Open
Abstract
The molecular machinery and chromosome structures carrying out meiosis are frequently conserved from yeast to mammals. However, signals initiating meiosis appear divergent: while nutrient restriction induces meiosis in the yeast system, retinoic acid (RA) and its target Stra8 have been shown to be necessary but not sufficient to induce meiotic initiation in mammalian germ cells. Here, we use primary culture of mouse undifferentiated spermatogonia without the support of gonadal somatic cells to show that nutrient restriction in combination with RA is sufficient to induce Stra8- and Spo11-dependent meiotic gene and chromosome programs that recapitulate the transcriptomic and cytologic features of in vivo meiosis. We demonstrate that neither nutrient restriction nor RA alone exerts these effects. Moreover, we identify a distinctive network of 11 nutrient restriction-upregulated transcription factor genes, which are associated with early meiosis in vivo and whose expression does not require RA. Our study proposes a conserved model, in which nutrient restriction induces meiotic initiation by upregulating key transcription factor genes for the meiotic gene program and provides an in vitro platform for meiotic induction that could facilitate research and haploid gamete production.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ning Wang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
18
|
Ribeiro J, Dupaigne P, Petrillo C, Ducrot C, Duquenne C, Veaute X, Saintomé C, Busso D, Guerois R, Martini E, Livera G. The meiosis-specific MEIOB-SPATA22 complex cooperates with RPA to form a compacted mixed MEIOB/SPATA22/RPA/ssDNA complex. DNA Repair (Amst) 2021; 102:103097. [PMID: 33812231 DOI: 10.1016/j.dnarep.2021.103097] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
During meiosis, programmed double-strand breaks are repaired by homologous recombination (HR) to form crossovers that are essential to homologous chromosome segregation. Single-stranded DNA (ssDNA) containing intermediates are key features of HR, which must be highly regulated. RPA, the ubiquitous ssDNA binding complex, was thought to play similar roles during mitotic and meiotic HR until the recent discovery of MEIOB and its partner, SPATA22, two essential meiosis-specific proteins. Here, we show that like MEIOB, SPATA22 resembles RPA subunits and binds ssDNA. We studied the physical and functional interactions existing between MEIOB, SPATA22, and RPA, and show that MEIOB and SPATA22 interact with the preformed RPA complex through their interacting domain and condense RPA-coated ssDNA in vitro. In meiotic cells, we show that MEIOB and SPATA22 modify the immunodetection of the two large subunits of RPA. Given these results, we propose that MEIOB-SPATA22 and RPA form a functional ssDNA-interacting complex to satisfy meiotic HR requirements by providing specific properties to the ssDNA.
Collapse
Affiliation(s)
- Jonathan Ribeiro
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France
| | - Pauline Dupaigne
- Laboratoire de Microscopie Moléculaire et Cellulaire, UMR 8126, Interactions Moléculaires et Cancer, CNRS, Université Paris Sud, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Cynthia Petrillo
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France
| | - Cécile Ducrot
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France
| | - Clotilde Duquenne
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France
| | - Xavier Veaute
- CIGEx, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA, Inserm, U1274, F-92260, Fontenay-aux-Roses, France
| | - Carole Saintomé
- MNHN, CNRS UMR 7196, INSERM U1154, Sorbonne Universités, 75231, Paris, France
| | - Didier Busso
- CIGEx, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA, Inserm, U1274, F-92260, Fontenay-aux-Roses, France
| | - Raphaël Guerois
- CNRS I2BC UMR 9198, iBiTec-S, SB²SM CEA SACLAY, 91191, Gif sur Yvette, France
| | - Emmanuelle Martini
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France.
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMR E008 Genetic Stability Stem Cells and Radiations, Université de Paris, Université Paris Saclay, CEA, F-92265, Fontenay aux Roses, France
| |
Collapse
|
19
|
Huang C, Guo T, Qin Y. Meiotic Recombination Defects and Premature Ovarian Insufficiency. Front Cell Dev Biol 2021; 9:652407. [PMID: 33763429 PMCID: PMC7982532 DOI: 10.3389/fcell.2021.652407] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Premature ovarian insufficiency (POI) is the depletion of ovarian function before 40 years of age due to insufficient oocyte formation or accelerated follicle atresia. Approximately 1–5% of women below 40 years old are affected by POI. The etiology of POI is heterogeneous, including genetic disorders, autoimmune diseases, infection, iatrogenic factors, and environmental toxins. Genetic factors account for 20–25% of patients. However, more than half of the patients were idiopathic. With the widespread application of next-generation sequencing (NGS), the genetic spectrum of POI has been expanded, especially the latest identification in meiosis and DNA repair-related genes. During meiotic prophase I, the key processes include DNA double-strand break (DSB) formation and subsequent homologous recombination (HR), which are essential for chromosome segregation at the first meiotic division and genome diversity of oocytes. Many animal models with defective meiotic recombination present with meiotic arrest, DSB accumulation, and oocyte apoptosis, which are similar to human POI phenotype. In the article, based on different stages of meiotic recombination, including DSB formation, DSB end processing, single-strand invasion, intermediate processing, recombination, and resolution and essential proteins involved in synaptonemal complex (SC), cohesion complex, and fanconi anemia (FA) pathway, we reviewed the individual gene mutations identified in POI patients and the potential candidate genes for POI pathogenesis, which will shed new light on the genetic architecture of POI and facilitate risk prediction, ovarian protection, and early intervention for POI women.
Collapse
Affiliation(s)
- Chengzi Huang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| | - Ting Guo
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| |
Collapse
|
20
|
Guo R, Xu Y, Leu NA, Zhang L, Fuchs SY, Ye L, Wang P. The ssDNA-binding protein MEIOB acts as a dosage-sensitive regulator of meiotic recombination. Nucleic Acids Res 2020; 48:12219-12233. [PMID: 33166385 PMCID: PMC7708077 DOI: 10.1093/nar/gkaa1016] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Meiotic recombination enables reciprocal exchange of genetic information between parental chromosomes and is essential for fertility. MEIOB, a meiosis-specific ssDNA-binding protein, regulates early meiotic recombination. Here we report that the human infertility-associated missense mutation (N64I) in MEIOB causes protein degradation and reduced crossover formation in mouse testes. Although the MEIOB N64I substitution is associated with human infertility, the point mutant mice are fertile despite meiotic defects. Meiob mutagenesis identifies serine 67 as a critical residue for MEIOB. Biochemically, these two mutations (N64I and S67 deletion) cause self-aggregation of MEIOB and sharply reduced protein half-life. Molecular genetic analyses of both point mutants reveal an important role for MEIOB in crossover formation in late meiotic recombination. Furthermore, we find that the MEIOB protein levels directly correlate with the severity of meiotic defects. Our results demonstrate that MEIOB regulates meiotic recombination in a dosage-dependent manner.
Collapse
Affiliation(s)
- Rui Guo
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yang Xu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - N Adrian Leu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Lei Zhang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Lan Ye
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
The novel male meiosis recombination regulator coordinates the progression of meiosis prophase I. J Genet Genomics 2020; 47:451-465. [PMID: 33250349 DOI: 10.1016/j.jgg.2020.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022]
Abstract
Meiosis is a specialized cell division for producing haploid gametes in sexually reproducing organisms. In this study, we have independently identified a novel meiosis protein male meiosis recombination regulator (MAMERR)/4930432K21Rik and showed that it is indispensable for meiosis prophase I progression in male mice. Using super-resolution structured illumination microscopy, we found that MAMERR functions at the same double-strand breaks as the replication protein A and meiosis-specific with OB domains/spermatogenesis associated 22 complex. We generated a Mamerr-deficient mouse model by deleting exons 3-6 and found that most of Mamerr-/- spermatocytes were arrested at pachynema and failed to progress to diplonema, although they exhibited almost intact synapsis and progression to the pachytene stage along with XY body formation. Further mechanistic studies revealed that the recruitment of DMC1/RAD51 and heat shock factor 2-binding protein in Mamerr-/- spermatocytes was only mildly impaired with a partial reduction in double-strand break repair, whereas a substantial reduction in ubiquitination on the autosomal axes and on the XY body appeared as a major phenotype in Mamerr-/- spermatocytes. We propose that MAMERR may participate in meiotic prophase I progression by regulating the ubiquitination of key meiotic proteins on autosomes and XY chromosomes, and in the absence of MAMERR, the repressed ubiquitination of key meiotic proteins leads to pachytene arrest and cell death.
Collapse
|
22
|
Paes VM, de Figueiredo JR, Ryan PL, Willard ST, Feugang JM. Comparative Analysis of Porcine Follicular Fluid Proteomes of Small and Large Ovarian Follicles. BIOLOGY 2020; 9:biology9050101. [PMID: 32429601 PMCID: PMC7285177 DOI: 10.3390/biology9050101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Ovarian follicular fluid is widely used for in vitro oocyte maturation, but its in-depth characterization to extract full beneficial effects remains unclear. Here, we performed both shotgun (nanoscale liquid chromatography coupled to tandem mass spectrometry or nanoLC-MS/MS) and gel-based (two dimension-differential in-gel electrophoresis or 2D-DIGE) proteomics, followed by functional bioinformatics to compare the proteomes of follicular fluids collected from small (<4 mm) and large (>6-12 mm) follicles of pig ovaries. A total of 2321 unique spots were detected with the 2D-DIGE across small and large follicles, while 2876 proteins with 88% successful annotations were detected with the shotgun approach. The shotgun and 2D-DIGE approaches revealed about 426 and 300 proteins that were respectively common across samples. Six proteins detected with both technical approaches were significantly differently expressed between small and large follicles. Pathways such as estrogen and PI3K-Akt signaling were significantly enriched in small follicles while the complement and coagulation cascades pathways were significantly represented in large follicles. Up-regulated proteins in small follicles were in favor of oocyte maturation, while those in large follicles were involved in the ovulatory process preparation. Few proteins with potential roles during sperm-oocyte interactions were especially detected in FF of large follicles and supporting the potential role of the ovarian FF on the intrafallopian sperm migration and interaction with the oocyte.
Collapse
Affiliation(s)
- Victor M. Paes
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA; (V.M.P.); (P.L.R.); (S.T.W.)
- Laboratory of Manipulation of Oocyte and Preantral follicles, State University of Ceará, CEP, 60740 903 Fortaleza, Brazil;
| | - José R. de Figueiredo
- Laboratory of Manipulation of Oocyte and Preantral follicles, State University of Ceará, CEP, 60740 903 Fortaleza, Brazil;
| | - Peter L. Ryan
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA; (V.M.P.); (P.L.R.); (S.T.W.)
| | - Scott T. Willard
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA; (V.M.P.); (P.L.R.); (S.T.W.)
| | - Jean M. Feugang
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA; (V.M.P.); (P.L.R.); (S.T.W.)
- Correspondence: ; Tel.: +662-325-7567; Fax: +662-325-8873
| |
Collapse
|
23
|
Petrillo C, Barroca V, Ribeiro J, Lailler N, Livera G, Keeney S, Martini E, Jain D. shani mutation in mouse affects splicing of Spata22 and leads to impaired meiotic recombination. Chromosoma 2020; 129:161-179. [PMID: 32388826 DOI: 10.1007/s00412-020-00735-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/14/2020] [Accepted: 04/26/2020] [Indexed: 02/07/2023]
Abstract
Recombination is crucial for chromosome pairing and segregation during meiosis. SPATA22, along with its direct binding partner and functional collaborator, MEIOB, is essential for the proper repair of double-strand breaks (DSBs) during meiotic recombination. Here, we describe a novel point-mutated allele (shani) of mouse Spata22 that we isolated in a forward genetic screen. shani mutant mice phenocopy Spata22-null and Meiob-null mice: mutant cells appear to form DSBs and initiate meiotic recombination, but are unable to complete DSB repair, leading to meiotic prophase arrest, apoptosis and sterility. shani mutants show precocious loss of DMC1 foci and improper accumulation of BLM-positive recombination foci, reinforcing the requirement of SPATA22-MEIOB for the proper progression of meiotic recombination events. The shani mutation lies within a Spata22 coding exon and molecular characterization shows that it leads to incorrect splicing of the Spata22 mRNA, ultimately resulting in no detectable SPATA22 protein. We propose that the shani mutation alters an exonic splicing enhancer element (ESE) within the Spata22 transcript. The affected DNA nucleotide is conserved in most tetrapods examined, suggesting that the splicing regulation we describe here may be a conserved feature of Spata22 regulation.
Collapse
Affiliation(s)
- Cynthia Petrillo
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem cells and Radiations, Université de Paris, Université Paris-Saclay, CEA, 92265, Fontenay aux Roses, France
| | - Vilma Barroca
- UMRE008 Genetic Stability Stem cells and Radiations, Université de Paris, Université Paris-Saclay, CEA, Inserm, U1274, 92265, Fontenay-aux-Roses, France
| | - Jonathan Ribeiro
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem cells and Radiations, Université de Paris, Université Paris-Saclay, CEA, 92265, Fontenay aux Roses, France
| | - Nathalie Lailler
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem cells and Radiations, Université de Paris, Université Paris-Saclay, CEA, 92265, Fontenay aux Roses, France
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Emmanuelle Martini
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem cells and Radiations, Université de Paris, Université Paris-Saclay, CEA, 92265, Fontenay aux Roses, France.
| | - Devanshi Jain
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
24
|
Xu Y, Liu R, Leu NA, Zhang L, Ibragmova I, Schultz DC, Wang PJ. A cell-based high-content screen identifies isocotoin as a small molecule inhibitor of the meiosis-specific MEIOB-SPATA22 complex†. Biol Reprod 2020; 103:333-342. [PMID: 32463099 PMCID: PMC7523692 DOI: 10.1093/biolre/ioaa062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/06/2020] [Accepted: 04/22/2020] [Indexed: 01/17/2023] Open
Abstract
MEIOB and SPATA22 are meiosis-specific proteins, interact with each other, and are essential for meiotic recombination and fertility. Aspartic acid 383 (D383) in MEIOB is critical for its interaction with SPATA22 in biochemical studies. Here we report that genetic studies validate the requirement of D383 for the function of MEIOB in mice. The MeiobD383A/D383A mice display meiotic arrest due to depletion of both MEIOB and SPATA22 proteins in the testes. We developed a cell-based bimolecular fluorescence complementation (BiFC) assay, in which MEIOB and SPATA22 are fused to split YFP moieties and their co-expression in cultured cells leads to the MEIOB–SPATA22 dimerization and reconstitution of the fluorophore. As expected, the interaction-disrupting D383A substitution results in the absence of YFP fluorescence in the BiFC assay. A high-throughput screen of small molecule libraries identified candidate hit compounds at a rate of 0.7%. Isocotoin, a hit compound from the natural product library, inhibits the MEIOB–SPATA22 interaction and promotes their degradation in HEK293 cells in a dose-dependent manner. Therefore, the BiFC assay can be employed to screen for small molecule inhibitors that disrupt protein–protein interactions or promote degradation of meiosis-specific proteins.
Collapse
Affiliation(s)
- Yang Xu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Rong Liu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA.,School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| | - N Adrian Leu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Lei Zhang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Ilsiya Ibragmova
- High-Throughput Screening Core, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David C Schultz
- High-Throughput Screening Core, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Kent K, Johnston M, Strump N, Garcia TX. Toward Development of the Male Pill: A Decade of Potential Non-hormonal Contraceptive Targets. Front Cell Dev Biol 2020; 8:61. [PMID: 32161754 PMCID: PMC7054227 DOI: 10.3389/fcell.2020.00061] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
With the continued steep rise of the global human population, and the paucity of safe and practical contraceptive options available to men, the need for development of effective and reversible non-hormonal methods of male fertility control is widely recognized. Currently there are several contraceptive options available to men, however, none of the non-hormonal alternatives have been clinically approved. To advance progress in the development of a safe and reversible contraceptive for men, further identification of novel reproductive tract-specific druggable protein targets is required. Here we provide an overview of genes/proteins identified in the last decade as specific or highly expressed in the male reproductive tract, with deletion phenotypes leading to complete male infertility in mice. These phenotypes include arrest of spermatogenesis and/or spermiogenesis, abnormal spermiation, abnormal spermatid morphology, abnormal sperm motility, azoospermia, globozoospermia, asthenozoospermia, and/or teratozoospermia, which are all desirable outcomes for a novel male contraceptive. We also consider other associated deletion phenotypes that could impact the desirability of a potential contraceptive. We further discuss novel contraceptive targets underscoring promising leads with the objective of presenting data for potential druggability and whether collateral effects may exist from paralogs with close sequence similarity.
Collapse
Affiliation(s)
- Katarzyna Kent
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States.,Department of Biology and Biotechnology, University of Houston-Clear Lake, Houston, TX, United States.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Madelaine Johnston
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Natasha Strump
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Thomas X Garcia
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States.,Department of Biology and Biotechnology, University of Houston-Clear Lake, Houston, TX, United States.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
26
|
Dapper AL, Payseur BA. Molecular evolution of the meiotic recombination pathway in mammals. Evolution 2019; 73:2368-2389. [PMID: 31579931 DOI: 10.1111/evo.13850] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
Meiotic recombination shapes evolution and helps to ensure proper chromosome segregation in most species that reproduce sexually. Recombination itself evolves, with species showing considerable divergence in the rate of crossing-over. However, the genetic basis of this divergence is poorly understood. Recombination events are produced via a complicated, but increasingly well-described, cellular pathway. We apply a phylogenetic comparative approach to a carefully selected panel of genes involved in the processes leading to crossovers-spanning double-strand break formation, strand invasion, the crossover/non-crossover decision, and resolution-to reconstruct the evolution of the recombination pathway in eutherian mammals and identify components of the pathway likely to contribute to divergence between species. Eleven recombination genes, predominantly involved in the stabilization of homologous pairing and the crossover/non-crossover decision, show evidence of rapid evolution and positive selection across mammals. We highlight TEX11 and associated genes involved in the synaptonemal complex and the early stages of the crossover/non-crossover decision as candidates for the evolution of recombination rate. Evolutionary comparisons to MLH1 count, a surrogate for the number of crossovers, reveal a positive correlation between genome-wide recombination rate and the rate of evolution at TEX11 across the mammalian phylogeny. Our results illustrate the power of viewing the evolution of recombination from a pathway perspective.
Collapse
Affiliation(s)
- Amy L Dapper
- Laboratory of Genetics, University of Wisconsin, Madison, Wisconsin, 53706.,Department of Biological Sciences, Mississippi State University, Mississippi, 39762
| | - Bret A Payseur
- Laboratory of Genetics, University of Wisconsin, Madison, Wisconsin, 53706
| |
Collapse
|
27
|
Pandey A, Yadav SK, Vishvkarma R, Singh B, Maikhuri JP, Rajender S, Gupta G. The dynamics of gene expression during and post meiosis sets the sperm agenda. Mol Reprod Dev 2019; 86:1921-1939. [DOI: 10.1002/mrd.23278] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Aastha Pandey
- Division of EndocrinologyCSIR‐Central Drug Research Institute Lucknow India
| | | | - Rahul Vishvkarma
- Division of EndocrinologyCSIR‐Central Drug Research Institute Lucknow India
| | - Bineta Singh
- Division of EndocrinologyCSIR‐Central Drug Research Institute Lucknow India
| | | | - Singh Rajender
- Division of EndocrinologyCSIR‐Central Drug Research Institute Lucknow India
| | - Gopal Gupta
- Division of EndocrinologyCSIR‐Central Drug Research Institute Lucknow India
| |
Collapse
|
28
|
Zhang Q, Ji SY, Busayavalasa K, Shao J, Yu C. Meiosis I progression in spermatogenesis requires a type of testis-specific 20S core proteasome. Nat Commun 2019; 10:3387. [PMID: 31358751 PMCID: PMC6662770 DOI: 10.1038/s41467-019-11346-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 07/05/2019] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is tightly regulated by ubiquitination and proteasomal degradation, especially during spermiogenesis, in which histones are replaced by protamine. However, the functions of proteasomal activity in meiosis I and II remain elusive. Here, we show that PSMA8-associated proteasomes are essential for the degradation of meiotic proteins and the progression of meiosis I during spermatogenesis. PSMA8 is expressed in spermatocytes from the pachytene stage, and assembles a type of testis-specific core proteasome. Deletion of PSMA8 decreases the abundance of proteasome in testes. Meiotic proteins that are normally degraded at late prophase I, such as RAD51 and RPA1, remain stable in PSMA8-deleted spermatocytes. Moreover, PSMA8-null spermatocytes exhibit delayed M-phase entry and are finally arrested at this stage, resulting in male infertility. However, PSMA8 is neither expressed nor required for female meiotic progression. Thus, meiosis I progression in spermatogenesis, particularly entry into and exit from M-phase, requires the proteasomal activity of PSMA8-associated proteasomes. Proteasomal degradation is required for the progression of spermatogenesis. Here the authors demonstrate that deletion of the testis-specific proteasome subunit PMSA8 leads to stabilization of the meiotic proteins RAD51 and RPA1 and a spermatogenic block at M-phase of meiosis I.
Collapse
Affiliation(s)
- Qianting Zhang
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, SE-40530, Sweden
| | - Shu-Yan Ji
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Kiran Busayavalasa
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, SE-40530, Sweden
| | - Jingchen Shao
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, SE-40530, Sweden
| | - Chao Yu
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, SE-40530, Sweden.
| |
Collapse
|
29
|
Palmer N, Talib SZA, Ratnacaram CK, Low D, Bisteau X, Lee JHS, Pfeiffenberger E, Wollmann H, Tan JHL, Wee S, Sobota R, Gunaratne J, Messerschmidt DM, Guccione E, Kaldis P. CDK2 regulates the NRF1/ Ehmt1 axis during meiotic prophase I. J Cell Biol 2019; 218:2896-2918. [PMID: 31350280 PMCID: PMC6719441 DOI: 10.1083/jcb.201903125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/10/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
Palmer et al. identify NRF1 as a novel CDK2 interactor and substrate. This interaction was found to be important for the DNA-binding activity of NRF1. Their findings demonstrate that the loss of CDK2 expression impairs the regulation of NRF1 transcriptional activity, leading to inappropriate transcription during meiotic division. Meiosis generates four genetically distinct haploid gametes over the course of two reductional cell divisions. Meiotic divisions are characterized by the coordinated deposition and removal of various epigenetic marks. Here we propose that nuclear respiratory factor 1 (NRF1) regulates transcription of euchromatic histone methyltransferase 1 (EHMT1) to ensure normal patterns of H3K9 methylation during meiotic prophase I. We demonstrate that cyclin-dependent kinase (CDK2) can bind to the promoters of a number of genes in male germ cells including that of Ehmt1 through interaction with the NRF1 transcription factor. Our data indicate that CDK2-mediated phosphorylation of NRF1 can occur at two distinct serine residues and negatively regulates NRF1 DNA binding activity in vitro. Furthermore, induced deletion of Cdk2 in spermatocytes results in increased expression of many NRF1 target genes including Ehmt1. We hypothesize that the regulation of NRF1 transcriptional activity by CDK2 may allow the modulation of Ehmt1 expression, therefore controlling the dynamic methylation of H3K9 during meiotic prophase.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Biochemistry, National University of Singapore, Singapore
| | - S Zakiah A Talib
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | | | - Diana Low
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Xavier Bisteau
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Joanna Hui Si Lee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | | | - Heike Wollmann
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Joel Heng Loong Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Biochemistry, National University of Singapore, Singapore
| | - Sheena Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Radoslaw Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Daniel M Messerschmidt
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Ernesto Guccione
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore .,Department of Biochemistry, National University of Singapore, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore .,Department of Biochemistry, National University of Singapore, Singapore
| |
Collapse
|
30
|
Zhang Q, Ji SY, Busayavalasa K, Yu C. SPO16 binds SHOC1 to promote homologous recombination and crossing-over in meiotic prophase I. SCIENCE ADVANCES 2019; 5:eaau9780. [PMID: 30746471 PMCID: PMC6357729 DOI: 10.1126/sciadv.aau9780] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/10/2018] [Indexed: 05/06/2023]
Abstract
Segregation of homologous chromosomes in meiosis I is tightly regulated by their physical links, or crossovers (COs), generated from DNA double-strand breaks (DSBs) through meiotic homologous recombination. In budding yeast, three ZMM (Zip1/2/3/4, Mer3, Msh4/5) proteins, Zip2, Zip4, and Spo16, form a "ZZS" complex, functioning to promote meiotic recombination via a DSB repair pathway. Here, we identified the mammalian ortholog of Spo16, termed SPO16, which interacts with the mammalian ortholog of Zip2 (SHOC1/MZIP2), and whose functions are evolutionarily conserved to promote the formation of COs. SPO16 localizes to the recombination nodules, as SHOC1 and TEX11 do. SPO16 is required for stabilization of SHOC1 and proper localization of other ZMM proteins. The DSBs formed in SPO16-deleted meiocytes were repaired without COs formation, although synapsis is less affected. Therefore, formation of SPO16-SHOC1 complex-associated recombination intermediates is a key step facilitating meiotic recombination that produces COs from yeast to mammals.
Collapse
Affiliation(s)
- Qianting Zhang
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Shu-Yan Ji
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Kiran Busayavalasa
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Chao Yu
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
- Corresponding author.
| |
Collapse
|
31
|
Zhang Q, Shao J, Fan HY, Yu C. Evolutionarily-conserved MZIP2 is essential for crossover formation in mammalian meiosis. Commun Biol 2018; 1:147. [PMID: 30272023 PMCID: PMC6155065 DOI: 10.1038/s42003-018-0154-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/30/2018] [Indexed: 01/14/2023] Open
Abstract
During meiosis, formation of crossovers-the physical links that ensure the segregation of homologous chromosomes-requires a group of evolutionarily conserved ZMM proteins. In budding yeast, three ZMM proteins, Zip2, Spo16, and Zip4, form a trimeric complex to bind recombination intermediates and promote crossover formation. Here, we show that MZIP2 is the mammalian ortholog of Zip2. Complete ablation of MZIP2 in mice caused sterility in both males and females, as well as defects in repairing meiotic DNA double-strand breaks. MZIP2 forms discrete foci on chromosomes axes, and is required for the localization of TEX11 (mammalian Zip4 ortholog) and another ZMM protein, MSH4, to form crossover-prone recombination intermediates. As a consequence, formation of crossovers is abolished and formation of synaptonemal complex is incomplete in MZIP2-null meiocytes, resulting in meiosis arrest at a zygotene-like stage. Our results suggest that the processing of early recombination intermediates toward mature crossovers is dependent on MZIP2.
Collapse
Affiliation(s)
- Qianting Zhang
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Jingchen Shao
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Chao Yu
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| |
Collapse
|
32
|
Monsef L, Borjian Boroujeni P, Totonchi M, Sabbaghian M, Mohseni Meybodi A. Gene alterations and expression spectrum of SPATA33 in nonobstructive azoospermic Iranian men. Mol Reprod Dev 2018; 85:760-767. [PMID: 30098056 DOI: 10.1002/mrd.23051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/09/2018] [Indexed: 01/26/2023]
Abstract
Genetic abnormalities have been considered a significant cause of male infertility. Increased expression of SPATA33 during the first wave of spermatogenesis indicates its possible association with the meiotic process. The aim of the current study was to investigate the genetic variations in the SPATA33 gene and its expression in patients with nonobstructive azoospermia (NOA). A total of 100 Iranian NOA men with idiopathic infertility were taken as the case group. The control group comprised 100 fertile men who had at least one child. The presence of nucleotide variations was analyzed in both groups using the standard polymerase chain reaction (PCR) sequencing technique. For mRNA and protein expression studies, testicular biopsy specimens from 27 patients were subdivided into three groups: nine obstructive azoospermic patients with hypospermatogenesis as control; nine maturation arrest (MA) and nine Sertoli cell-only syndromes (SCOS) as case groups. The expression of SPATA33 at both mRNA and protein levels was compared among these three groups using the reverse transcription PCR technique, the realtime-PCR technique, and immunohistochemistry. Mutation analysis of the SPATA33 gene revealed five nucleotide changes among the population studied. All but one showed no significant differences between the groups. The genotype distributions of rs112536073A > T in the transcription factor binding site region with heterozygote and homozygote genotypes were significantly different ( p < 0.05) between the two groups. More heterozygotes of this polymorphism were observed in patients, whereas more homozygotes were detected in controls. Accordingly, the current study illustrated that alterations in SPATA33 gene, at least those found in this study, may not impair spermatogenesis in patients with NOA. Reduction of gene expression at the level of mRNA in patients with SCOS can be interpreted by the absence of germ cells in the testicular tissue of these patients.
Collapse
Affiliation(s)
- Ladan Monsef
- Department of Basic Science and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Parnaz Borjian Boroujeni
- Department of Genetics, Reproductive Biomedicine 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
| | - Marjan Sabbaghian
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Anahita Mohseni Meybodi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
33
|
Xu Y, Greenberg RA, Schonbrunn E, Wang PJ. Meiosis-specific proteins MEIOB and SPATA22 cooperatively associate with the single-stranded DNA-binding replication protein A complex and DNA double-strand breaks. Biol Reprod 2018; 96:1096-1104. [PMID: 28453612 DOI: 10.1093/biolre/iox040] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/26/2017] [Indexed: 12/27/2022] Open
Abstract
Meiotic recombination ensures faithful segregation of homologous chromosomes during meiosis and generates genetic diversity in gametes. MEIOB (meiosis specific with OB domains), a meiosis-specific single-stranded DNA-binding homolog of replication protein A1 (RPA1), is essential for meiotic recombination. Here, we investigated the molecular mechanisms of MEIOB by characterizing its binding partners spermatogenesis associated 22 (SPATA22) and RPA. We find that MEIOB and SPATA22 form an obligate complex and contain defined interaction domains. The interaction between these two proteins is unusual in that nearly any deletion in the binding domains abolishes the interaction. Strikingly, a single residue D383 in MEIOB is indispensable for the interaction. The MEIOB/SPATA22 complex interacts with the RPA heterotrimeric complex in a collaborative manner. Furthermore, MEIOB and SPATA22 are recruited to induced DNA double-strand breaks (DSBs) together but not alone. These results demonstrate the cooperative property of the MEIOB-SPATA22 complex in its interaction with RPA and recruitment to DSBs.
Collapse
Affiliation(s)
- Yang Xu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Roger A Greenberg
- Departments of Cancer Biology and Pathology, Abramson Family Cancer Research Institute, Basser Research Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ernst Schonbrunn
- Drug Discovery Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Unpackaging the genetics of mammalian fertility: strategies to identify the “reproductive genome”†. Biol Reprod 2018; 99:1119-1128. [DOI: 10.1093/biolre/ioy133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/05/2018] [Indexed: 12/18/2022] Open
|
35
|
Liu XM, Wang YK, Liu YH, Yu XX, Wang PC, Li X, Du ZQ, Yang CX. Single-cell transcriptome sequencing reveals that cell division cycle 5-like protein is essential for porcine oocyte maturation. J Biol Chem 2017; 293:1767-1780. [PMID: 29222335 DOI: 10.1074/jbc.m117.809608] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/03/2017] [Indexed: 02/02/2023] Open
Abstract
The brilliant cresyl blue (BCB) test is used in both basic biological research and assisted reproduction to identify oocytes likely to be developmentally competent. However, the underlying molecular mechanism targeted by the BCB test is still unclear. To explore this question, we first confirmed that BCB-positive porcine oocytes had higher rates of meiotic maturation, better rates of cleavage and development into blastocysts, and lower death rates. Subsequent single-cell transcriptome sequencing on porcine germinal vesicle (GV)-stage oocytes identified 155 genes that were significantly differentially expressed between BCB-negative and BCB-positive oocytes. These included genes such as cdc5l, ldha, spata22, rgs2, paip1, wee1b, and hsp27, which are enriched in functionally important signaling pathways including cell cycle regulation, oocyte meiosis, spliceosome formation, and nucleotide excision repair. In BCB-positive GV oocytes that additionally had a lower frequency of DNA double-strand breaks, the CDC5L protein was significantly more abundant. cdc5l/CDC5L inhibition by short interference (si)RNA or antibody microinjection significantly impaired porcine oocyte meiotic maturation and subsequent parthenote development. Taken together, our single-oocyte sequencing data point to a potential new role for CDC5L in porcine oocyte meiosis and early embryo development, and supports further analysis of this protein in the context of the BCB test.
Collapse
Affiliation(s)
- Xiao-Man Liu
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yan-Kui Wang
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yun-Hua Liu
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiao-Xia Yu
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Pei-Chao Wang
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xuan Li
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Zhi-Qiang Du
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Cai-Xia Yang
- From the Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
36
|
Hays E, Majchrzak N, Daniel V, Ferguson Z, Brown S, Hathorne K, La Salle S. Spermatogenesis associated 22 is required for DNA repair and synapsis of homologous chromosomes in mouse germ cells. Andrology 2017; 5:299-312. [PMID: 28297563 DOI: 10.1111/andr.12315] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/12/2016] [Accepted: 11/16/2016] [Indexed: 01/09/2023]
Abstract
Analysis of the N-ethyl-N-nitrosourea (ENU)-induced repro42 mutation previously identified spermatogenesis associated 22 (Spata22) as a gene required for meiotic progression and fertility in both male and female mice, but its specific contribution to the process was unclear. Here, we report on a novel, null allele of Spata22 (Spata22Gt ) and confirm its requirement for germ cell development. Similar to repro42 mutant mice, histological and mating analyses indicate that gametogenesis is profoundly affected in Spata22Gt/Gt males and females, resulting in infertility. Cytological examination confirms that germ cells do not progress beyond zygonema and meiotic arrest is linked to impairment of both synapsis and DNA repair. Analysis of SPATA22 distribution reveals that it localizes to foci associated with meiotic chromosomes during prophase I and that the number of foci peaks at zygonema; there are also more SPATA22 foci in oocytes than in spermatocytes. Furthermore, SPATA22 co-localizes with a number of proteins involved in meiotic recombination, including RAD51, DMC1, and MLH1, and is present until mid-pachynema, suggesting a role in resolution of recombination intermediates. In fact, SPATA22 co-localizes with MLH1 in more than 20% of foci at pachynema. Analysis of Spata22Gt/Gt meiocytes confirms that SPATA22 is required for localization of MEIOB but not RPA (two proteins known to interact with SPATA22), and immunoblotting corroborates that production of MEIOB is indeed decreased in the absence of SPATA22. Together, these data suggest that SPATA22 is required for both meiotic recombination and synapsis during meiosis in mice.
Collapse
Affiliation(s)
- E Hays
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - N Majchrzak
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA
| | - V Daniel
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - Z Ferguson
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, Downers Grove, IL, USA
| | - S Brown
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, Downers Grove, IL, USA
| | - K Hathorne
- Department of Biomedical Sciences, College of Health Sciences, Midwestern University, Downers Grove, IL, USA
| | - S La Salle
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| |
Collapse
|
37
|
Regulatory complexity revealed by integrated cytological and RNA-seq analyses of meiotic substages in mouse spermatocytes. BMC Genomics 2016; 17:628. [PMID: 27519264 PMCID: PMC4983049 DOI: 10.1186/s12864-016-2865-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/28/2016] [Indexed: 01/24/2023] Open
Abstract
Background The continuous and non-synchronous nature of postnatal male germ-cell development has impeded stage-specific resolution of molecular events of mammalian meiotic prophase in the testis. Here the juvenile onset of spermatogenesis in mice is analyzed by combining cytological and transcriptomic data in a novel computational analysis that allows decomposition of the transcriptional programs of spermatogonia and meiotic prophase substages. Results Germ cells from testes of individual mice were obtained at two-day intervals from 8 to 18 days post-partum (dpp), prepared as surface-spread chromatin and immunolabeled for meiotic stage-specific protein markers (STRA8, SYCP3, phosphorylated H2AFX, and HISTH1T). Eight stages were discriminated cytologically by combinatorial antibody labeling, and RNA-seq was performed on the same samples. Independent principal component analyses of cytological and transcriptomic data yielded similar patterns for both data types, providing strong evidence for substage-specific gene expression signatures. A novel permutation-based maximum covariance analysis (PMCA) was developed to map co-expressed transcripts to one or more of the eight meiotic prophase substages, thereby linking distinct molecular programs to cytologically defined cell states. Expression of meiosis-specific genes is not substage-limited, suggesting regulation of substage transitions at other levels. Conclusions This integrated analysis provides a general method for resolving complex cell populations. Here it revealed not only features of meiotic substage-specific gene expression, but also a network of substage-specific transcription factors and relationships to potential target genes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2865-1) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Miao H, Miao CX, Li N, Han J. FOXJ2 controls meiosis during spermatogenesis in male mice. Mol Reprod Dev 2016; 83:684-91. [PMID: 27316861 DOI: 10.1002/mrd.22671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/13/2016] [Indexed: 12/17/2022]
Abstract
Spermatogenesis is a highly complex cell differentiation process necessary for production of haploid spermatozoa. Central to this unique process is spermatocyte meiosis. FOXJ2 (Forkhead box J2), a FOX transcription factor, is specifically expressed in meiotic spermatocytes in adult mouse testes, so we used a germ cell specific conditional knockout model (Foxj2(flox/flox) , Mvh-Cre) to explore its role in spermatogenesis. Loss of FOXJ2 in the male germ line led to meiotic arrest and complete infertility. Although, DNA double-strand breaks (DSBs) were initiated, Foxj2-deficient spermatocytes failed to form chromosomal synapses and perform DSB repair. Furthermore, Foxj2-deficient spermatocytes contained significantly less mRNA encoding DSB repair-associated factors (Rad18, Rad51, Brca1, Brca2, and Tex15) and meiotic arrest-related proteins (Fzr1, Hsp70-2, Spata22, Eif4g3, and Zpac); in contrast, no change was observed in the expression of spermatogonia markers (Gfra1, Zbtb16, and c-Kit) and germ cell markers (Dazl, Mvh, and Tra98). Taken together, FOXJ2 appears to promote meiotic progression in male mice by a mechanism that needs further investigation. Mol. Reprod. Dev. 83: 684-691, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hui Miao
- Department of, Reproduction and Genetics, ChangZhi Medical College Affiliated HePing Hospital, ShanXi Province, China
| | - Cong-Xiu Miao
- Department of, Reproduction and Genetics, ChangZhi Medical College Affiliated HePing Hospital, ShanXi Province, China
| | - Na Li
- Department of, Reproduction and Genetics, ChangZhi Medical College Affiliated HePing Hospital, ShanXi Province, China
| | - Jing Han
- Department of, Reproduction and Genetics, ChangZhi Medical College Affiliated HePing Hospital, ShanXi Province, China
| |
Collapse
|
39
|
Abby E, Tourpin S, Ribeiro J, Daniel K, Messiaen S, Moison D, Guerquin J, Gaillard JC, Armengaud J, Langa F, Toth A, Martini E, Livera G. Implementation of meiosis prophase I programme requires a conserved retinoid-independent stabilizer of meiotic transcripts. Nat Commun 2016; 7:10324. [PMID: 26742488 PMCID: PMC4729902 DOI: 10.1038/ncomms10324] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/27/2015] [Indexed: 12/28/2022] Open
Abstract
Sexual reproduction is crucially dependent on meiosis, a conserved, specialized cell division programme that is essential for the production of haploid gametes. Here we demonstrate that fertility and the implementation of the meiotic programme require a previously uncharacterized meiosis-specific protein, MEIOC. Meioc invalidation in mice induces early and pleiotropic meiotic defects in males and females. MEIOC prevents meiotic transcript degradation and interacts with an RNA helicase that binds numerous meiotic mRNAs. Our results indicate that proper engagement into meiosis necessitates the specific stabilization of meiotic transcripts, a previously little-appreciated feature in mammals. Remarkably, the upregulation of MEIOC at the onset of meiosis does not require retinoic acid and STRA8 signalling. Thus, we propose that the complete induction of the meiotic programme requires both retinoic acid-dependent and -independent mechanisms. The latter process involving post-transcriptional regulation likely represents an ancestral mechanism, given that MEIOC homologues are conserved throughout multicellular animals. Meiosis is a cell division program that produces haploid gametes and is initiated by a retinoic acid-dependent process. Here the authors report that a meiosis-specific protein, MEIOC, is upregulated in a retinoic acid-independent manner and is required to stabilise meiosis-specific transcripts.
Collapse
Affiliation(s)
- Emilie Abby
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Sophie Tourpin
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Jonathan Ribeiro
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Katrin Daniel
- Molecular Cell Biology Group/Experimental Center, Institute of Physiological Chemistry, Medical School, MTZ, Dresden University of Technology, Fiedlerstrasse 42, Dresden 01307, Germany
| | - Sébastien Messiaen
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Delphine Moison
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Justine Guerquin
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Jean-Charles Gaillard
- CEA, DSV/IBITEC-S/SPI/Li2D, Laboratory 'Innovative Technologies for Detection and Diagnostic', CEA-Marcoule, BP 17171, Bagnols-sur-Cèze F-30200, France
| | - Jean Armengaud
- CEA, DSV/IBITEC-S/SPI/Li2D, Laboratory 'Innovative Technologies for Detection and Diagnostic', CEA-Marcoule, BP 17171, Bagnols-sur-Cèze F-30200, France
| | - Francina Langa
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, Paris 75015, France
| | - Attila Toth
- Molecular Cell Biology Group/Experimental Center, Institute of Physiological Chemistry, Medical School, MTZ, Dresden University of Technology, Fiedlerstrasse 42, Dresden 01307, Germany
| | - Emmanuelle Martini
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Gabriel Livera
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| |
Collapse
|
40
|
Ribeiro J, Abby E, Livera G, Martini E. RPA homologs and ssDNA processing during meiotic recombination. Chromosoma 2015; 125:265-76. [PMID: 26520106 PMCID: PMC4830875 DOI: 10.1007/s00412-015-0552-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 09/12/2015] [Accepted: 10/14/2015] [Indexed: 11/25/2022]
Abstract
Meiotic homologous recombination is a specialized process that involves homologous chromosome pairing and strand exchange to guarantee proper chromosome segregation and genetic diversity. The formation and repair of DNA double-strand breaks (DSBs) during meiotic recombination differs from those during mitotic recombination in that the homologous chromosome rather than the sister chromatid is the preferred repair template. The processing of single-stranded DNA (ssDNA) formed on intermediate recombination structures is central to driving the specific outcomes of DSB repair during meiosis. Replication protein A (RPA) is the main ssDNA-binding protein complex involved in DNA metabolism. However, the existence of RPA orthologs in plants and the recent discovery of meiosis specific with OB domains (MEIOB), a widely conserved meiosis-specific RPA1 paralog, strongly suggest that multiple RPA complexes evolved and specialized to subdivide their roles during DNA metabolism. Here we review ssDNA formation and maturation during mitotic and meiotic recombination underlying the meiotic specific features. We describe and discuss the existence and properties of MEIOB and multiple RPA subunits in plants and highlight how they can provide meiosis-specific fates to ssDNA processing during homologous recombination. Understanding the functions of these RPA homologs and how they interact with the canonical RPA subunits is of major interest in the fields of meiosis and DNA repair.
Collapse
Affiliation(s)
- Jonathan Ribeiro
- Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, University of Paris Diderot, Sorbonne Paris Cité, UMR 967, F-92265, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, F-92265, Fontenay-aux-Roses, France
- INSERM, Unité 967, F-92265, Fontenay-aux-Roses, France
- Université Paris-Saclay, UMR-967, F-92265, Fontenay-aux-Roses, France
| | - Emilie Abby
- Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, University of Paris Diderot, Sorbonne Paris Cité, UMR 967, F-92265, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, F-92265, Fontenay-aux-Roses, France
- INSERM, Unité 967, F-92265, Fontenay-aux-Roses, France
- Université Paris-Saclay, UMR-967, F-92265, Fontenay-aux-Roses, France
| | - Gabriel Livera
- Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, University of Paris Diderot, Sorbonne Paris Cité, UMR 967, F-92265, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, F-92265, Fontenay-aux-Roses, France
- INSERM, Unité 967, F-92265, Fontenay-aux-Roses, France
- Université Paris-Saclay, UMR-967, F-92265, Fontenay-aux-Roses, France
| | - Emmanuelle Martini
- Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, University of Paris Diderot, Sorbonne Paris Cité, UMR 967, F-92265, Fontenay-aux-Roses, France.
- CEA, DSV, iRCM, SCSR, LDG, F-92265, Fontenay-aux-Roses, France.
- INSERM, Unité 967, F-92265, Fontenay-aux-Roses, France.
- Université Paris-Saclay, UMR-967, F-92265, Fontenay-aux-Roses, France.
| |
Collapse
|
41
|
Abstract
The study of homologous recombination has its historical roots in meiosis. In this context, recombination occurs as a programmed event that culminates in the formation of crossovers, which are essential for accurate chromosome segregation and create new combinations of parental alleles. Thus, meiotic recombination underlies both the independent assortment of parental chromosomes and genetic linkage. This review highlights the features of meiotic recombination that distinguish it from recombinational repair in somatic cells, and how the molecular processes of meiotic recombination are embedded and interdependent with the chromosome structures that characterize meiotic prophase. A more in-depth review presents our understanding of how crossover and noncrossover pathways of meiotic recombination are differentiated and regulated. The final section of this review summarizes the studies that have defined defective recombination as a leading cause of pregnancy loss and congenital disease in humans.
Collapse
Affiliation(s)
- Neil Hunter
- Howard Hughes Medical Institute, Department of Microbiology & Molecular Genetics, Department of Molecular & Cellular Biology, Department of Cell Biology & Human Anatomy, University of California Davis, Davis, California 95616
| |
Collapse
|
42
|
Sarver AE, Sarver AL, Thayanithy V, Subramanian S. Identification, by systematic RNA sequencing, of novel candidate biomarkers and therapeutic targets in human soft tissue tumors. J Transl Med 2015; 95:1077-88. [PMID: 26121316 DOI: 10.1038/labinvest.2015.80] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/17/2015] [Accepted: 05/11/2015] [Indexed: 01/14/2023] Open
Abstract
Human sarcomas comprise a heterogeneous group of more than 50 subtypes broadly classified into two groups: bone and soft tissue sarcomas. Such heterogeneity and their relative rarity have made them challenging targets for classification, biomarker identification, and development of improved treatment strategies. In this study, we used RNA sequencing to analyze 35 primary human tissue samples representing 13 different sarcoma subtypes, along with benign schwannoma, and normal bone and muscle tissues. For each sarcoma subtype, we detected unique messenger RNA (mRNA) expression signatures, which we further subjected to bioinformatic functional analysis, upstream regulatory analysis, and microRNA (miRNA) targeting analysis. We found that, for each sarcoma subtype, significantly upregulated genes and their deduced upstream regulators included not only previously implicated known players but also novel candidates not previously reported to be associated with sarcoma. For example, the schwannoma samples were characterized by high expression of not only the known associated proteins GFAP and GAP43 but also the novel player GJB6. Further, when we integrated our expression profiles with miRNA expression data from each sarcoma subtype, we were able to deduce potential key miRNA-gene regulator relationships for each. In the Ewing's sarcoma and fibromatosis samples, two sarcomas where miR-182-5p is significantly downregulated, multiple predicted targets were significantly upregulated, including HMCN1, NKX2-2, SCNN1G, and SOX2. In conclusion, despite the small number of samples per sarcoma subtype, we were able to identify key known players; concurrently, we discovered novel genes that may prove to be important in the molecular classification of sarcomas and in the development of novel treatments.
Collapse
Affiliation(s)
- Anne E Sarver
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Venugopal Thayanithy
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Subbaya Subramanian
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
43
|
Ohno Y, Shimizu S, Tatara A, Imaoku T, Ishii T, Sasa M, Serikawa T, Kuramoto T. Hcn1 is a tremorgenic genetic component in a rat model of essential tremor. PLoS One 2015; 10:e0123529. [PMID: 25970616 PMCID: PMC4430019 DOI: 10.1371/journal.pone.0123529] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/18/2015] [Indexed: 01/12/2023] Open
Abstract
Genetic factors are thought to play a major role in the etiology of essential tremor (ET); however, few genetic changes that induce ET have been identified to date. In the present study, to find genes responsible for the development of ET, we employed a rat model system consisting of a tremulous mutant strain, TRM/Kyo (TRM), and its substrain TRMR/Kyo (TRMR). The TRM rat is homozygous for the tremor (tm) mutation and shows spontaneous tremors resembling human ET. The TRMR rat also carries a homozygous tm mutation but shows no tremor, leading us to hypothesize that TRM rats carry one or more genes implicated in the development of ET in addition to the tm mutation. We used a positional cloning approach and found a missense mutation (c. 1061 C>T, p. A354V) in the hyperpolarization-activated cyclic nucleotide-gated 1 channel (Hcn1) gene. The A354V HCN1 failed to conduct hyperpolarization-activated currents in vitro, implicating it as a loss-of-function mutation. Blocking HCN1 channels with ZD7288 in vivo evoked kinetic tremors in nontremulous TRMR rats. We also found neuronal activation of the inferior olive (IO) in both ZD7288-treated TRMR and non-treated TRM rats and a reduced incidence of tremor in the IO-lesioned TRM rats, suggesting a critical role of the IO in tremorgenesis. A rat strain carrying the A354V mutation alone on a genetic background identical to that of the TRM rats showed no tremor. Together, these data indicate that body tremors emerge when the two mutant loci, tm and Hcn1A354V, are combined in a rat model of ET. In this model, HCN1 channels play an important role in the tremorgenesis of ET. We propose that oligogenic, most probably digenic, inheritance is responsible for the genetic heterogeneity of ET.
Collapse
Affiliation(s)
- Yukihiro Ohno
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Saki Shimizu
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Ayaka Tatara
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Takuji Imaoku
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
| | - Takahiro Ishii
- Department of Physiology and Neurobiology, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| | | | - Tadao Serikawa
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, 569–1094, Japan
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| | - Takashi Kuramoto
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, 606–8501, Japan
| |
Collapse
|
44
|
Abstract
Homology search and DNA strand-exchange reactions are central to homologous recombination in meiosis. During meiosis, these processes are regulated such that the probability of choosing a homolog chromatid as recombination partner is enhanced relative to that of choosing a sister chromatid. This regulatory process occurs as homologous chromosomes pair in preparation for assembly of the synaptonemal complex. Two strand-exchange proteins, Rad51 and Dmc1, cooperate in regulated homology search and strand exchange in most organisms. Here, we summarize studies on the properties of these two proteins and their accessory factors. In addition, we review current models for the assembly of meiotic strand-exchange complexes and the possible mechanisms through which the interhomolog bias of recombination partner choice is achieved.
Collapse
Affiliation(s)
- M Scott Brown
- Department of Radiation and Cellular Oncology, and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637
| | - Douglas K Bishop
- Department of Radiation and Cellular Oncology, and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
45
|
Genetic evidence suggests that Spata22 is required for the maintenance of Rad51 foci in mammalian meiosis. Sci Rep 2014; 4:6148. [PMID: 25142975 PMCID: PMC4139951 DOI: 10.1038/srep06148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 08/01/2014] [Indexed: 11/08/2022] Open
Abstract
Meiotic nodules are the sites of double-stranded DNA break repair. Rpa is a single-stranded DNA-binding protein, and Rad51 is a protein that assists in the repair of DNA double strand breaks. The localisation of Rad51 to meiotic nodules before the localisation of Rpa in mice introduces the issue of whether Rpa is involved in presynaptic filament formation during mammalian meiosis. Here, we show that a protein with unknown function, Spata22, colocalises with Rpa in meiotic nodules in rat spermatocytes. In spermatocytes of Spata22-deficient mutant rats, meiosis was arrested at the zygotene-like stage, and a normal number of Rpa foci was observed during leptotene- and zygotene-like stages. The number of Rad51 foci was initially normal but declined from the leptotene-like stage. These results suggest that both formation and maintenance of Rpa foci are independent of Spata22, and the maintenance, but not the formation, of Rad51 foci requires Spata22. We propose a possible model of presynaptic filament formation in mammalian meiosis, which involves Rpa and Spata22.
Collapse
|
46
|
Luo M, Yang F, Leu NA, Landaiche J, Handel MA, Benavente R, La Salle S, Wang PJ. MEIOB exhibits single-stranded DNA-binding and exonuclease activities and is essential for meiotic recombination. Nat Commun 2014; 4:2788. [PMID: 24240703 PMCID: PMC3891831 DOI: 10.1038/ncomms3788] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/16/2013] [Indexed: 12/21/2022] Open
Abstract
Meiotic recombination enables the reciprocal exchange of genetic material between parental homologous chromosomes, and ensures faithful chromosome segregation during meiosis in sexually reproducing organisms. This process relies on the complex interaction of DNA repair factors and many steps remain poorly understood in mammals. Here we report the identification of MEIOB, a meiosis-specific protein, in a proteomics screen for novel meiotic chromatin-associated proteins in mice. MEIOB contains an OB domain with homology to one of the RPA1 OB folds. MEIOB binds to single-stranded DNA and exhibits 3'-5' exonuclease activity. MEIOB forms a complex with RPA and with SPATA22, and these three proteins co-localize in foci that are associated with meiotic chromosomes. Strikingly, chromatin localization and stability of MEIOB depends on SPATA22 and vice versa. Meiob-null mouse mutants exhibit a failure in meiosis and sterility in both sexes. Our results suggest that MEIOB is required for meiotic recombination and chromosomal synapsis.
Collapse
Affiliation(s)
- Mengcheng Luo
- Center for Animal Transgenesis and Germ Cell Research, Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, 200E Old Vet, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lou Y, Liu W, Wang C, Huang L, Jin S, Lin Y, Zheng Y. Histological evaluation and Prdm9 expression level in the testis of sterile male cattle-yaks. Livest Sci 2014. [DOI: 10.1016/j.livsci.2013.12.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Ishishita S, Inui T, Matsuda Y, Serikawa T, Kitada K. Infertility associated with meiotic failure in the tremor rat (tm/tm) is caused by the deletion of spermatogenesis associated 22. Exp Anim 2014; 62:219-27. [PMID: 23903057 PMCID: PMC4160939 DOI: 10.1538/expanim.62.219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The tremor rat is an autosomal recessive mutant exhibiting sterility
with gonadal hypoplasia in both sexes. The causative mutation tremor
(tm) is known as a genomic deletion spanning >200 kb in Chr 10q24.
Spermatogenesis associated 22 (Spata22) has been shown
to be a vertebrate-specific gene essential for the progression of meiosis through prophase
I and completion of chromosome synapsis and meiotic recombination using a mouse
repro42 mutant carrying an
N-ethyl-N-nitrosourea (ENU)-induced nonsense mutation in
Spata22. In this study, we show that Spata22 was
identified as the gene responsible for the failure of gametogenesis to progress beyond
meiosis I in tm homozygous rats by a transgenic rescue experiment.
Meiosis was arrested during prophase I in the mutant testis. Precise mapping of the
breakage point revealed that the deleted genomic region spanned approximately 240 kb and
comprised at least 13 genes, including Spata22. Rat
Spata22 was predominantly expressed in the testis, and its
transcription increased with the first wave of spermatogenesis, as seen in the mouse
ortholog. These results suggest that Spata22 may play an important role
in meiotic prophase I in rats, as seen in mice, and that the tm
homozygous rat may be useful for investigating the physiological function of
Spata22, as an experimental system for clarifying the effect of a null
mutation, and may be an animal model for studying the pathogenesis and treatment of
infertility caused by impaired meiosis.
Collapse
Affiliation(s)
- Satoshi Ishishita
- Division of Bioscience, Graduate School of Environmental Earth Science, Hokkaido University, North 10 West 8, Kita-ku, Sapporo 060-0810, Japan
| | | | | | | | | |
Collapse
|
49
|
Margolin G, Khil PP, Kim J, Bellani MA, Camerini-Otero RD. Integrated transcriptome analysis of mouse spermatogenesis. BMC Genomics 2014; 15:39. [PMID: 24438502 PMCID: PMC3906902 DOI: 10.1186/1471-2164-15-39] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 01/14/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Differentiation of primordial germ cells into mature spermatozoa proceeds through multiple stages, one of the most important of which is meiosis. Meiotic recombination is in turn a key part of meiosis. To achieve the highly specialized and diverse functions necessary for the successful completion of meiosis and the generation of spermatozoa thousands of genes are coordinately regulated through spermatogenesis. A complete and unbiased characterization of the transcriptome dynamics of spermatogenesis is, however, still lacking. RESULTS In order to characterize gene expression during spermatogenesis we sequenced eight mRNA samples from testes of juvenile mice from 6 to 38 days post partum. Using gene expression clustering we defined over 1,000 novel meiotically-expressed genes. We also developed a computational de-convolution approach and used it to estimate cell type-specific gene expression in pre-meiotic, meiotic and post-meiotic cells. In addition, we detected 13,000 novel alternative splicing events around 40% of which preserve an open reading frame, and found experimental support for 159 computational gene predictions. A comparison of RNA polymerase II (Pol II) ChIP-Seq signals with RNA-Seq coverage shows that gene expression correlates well with Pol II signals, both at promoters and along the gene body. However, we observe numerous instances of non-canonical promoter usage, as well as intergenic Pol II peaks that potentially delineate unannotated promoters, enhancers or small RNA clusters. CONCLUSIONS Here we provide a comprehensive analysis of gene expression throughout mouse meiosis and spermatogenesis. Importantly, we find over a thousand of novel meiotic genes and over 5,000 novel potentially coding isoforms. These data should be a valuable resource for future studies of meiosis and spermatogenesis in mammals.
Collapse
Affiliation(s)
- Gennady Margolin
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Building 5, Room 205A, Bethesda, MD 20892, USA
| | - Pavel P Khil
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Building 5, Room 205A, Bethesda, MD 20892, USA
| | - Joongbaek Kim
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Building 5, Room 205A, Bethesda, MD 20892, USA
| | - Marina A Bellani
- National Institute of Aging, National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - R Daniel Camerini-Otero
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Building 5, Room 205A, Bethesda, MD 20892, USA
| |
Collapse
|
50
|
Baudat F, Imai Y, de Massy B. Meiotic recombination in mammals: localization and regulation. Nat Rev Genet 2013; 14:794-806. [PMID: 24136506 DOI: 10.1038/nrg3573] [Citation(s) in RCA: 398] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During meiosis, a programmed induction of DNA double-strand breaks (DSBs) leads to the exchange of genetic material between homologous chromosomes. These exchanges increase genome diversity and are essential for proper chromosome segregation at the first meiotic division. Recent findings have highlighted an unexpected molecular control of the distribution of meiotic DSBs in mammals by a rapidly evolving gene, PR domain-containing 9 (PRDM9), and genome-wide analyses have facilitated the characterization of meiotic DSB sites at unprecedented resolution. In addition, the identification of new players in DSB repair processes has allowed the delineation of recombination pathways that have two major outcomes, crossovers and non-crossovers, which have distinct mechanistic roles and consequences for genome evolution.
Collapse
Affiliation(s)
- Frédéric Baudat
- Institute of Human Genetics, Unité Propre de Recherche 1142, Centre National de la Recherche Scientifique, 141 rue de la Cardonille, 34396 Montpellier, France
| | | | | |
Collapse
|