1
|
Rosa IR, Barbosa CP, Ferrandez CA, Sonoda BDB, Christofolini DM, Trevisan CM, Laganà AS, Peluso C, Bianco B. Involvement of SYCP2L and TDRD3 gene variants on ovarian reserve and reproductive outcomes: a cross-sectional study. JBRA Assist Reprod 2023; 27:428-435. [PMID: 37417852 PMCID: PMC10712833 DOI: 10.5935/1518-0557.20220074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 12/26/2022] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVE Single nucleotide variants have been implicated in the response to fertility treatment and a pharmacogenomic approach may help to customize therapy based on patient genome. We aimed to evaluate the effect, individual and combined, of SYCP2L (rs2153157:G>A) and TDRD3 (rs4886238:G>A) variants on ovarian reserve, response to controlled ovarian stimulation (COS) and reproductive outcomes of women undergoing in vitro fertilization (IVF) treatment. METHODS This cross-sectional study included 149 normoovulatory women undergoing IVF. Genotyping was performed using the TaqMan real-time polymerase chain reaction method. Clinical parameters and reproductive outcomes were compared according to the genotypes of the variants studied. RESULTS Considering ovarian reserve, there were no significant differences among SYCP2L or TDRD3 genotypes in terms of FSH levels or AFC; however, AMH levels were significantly different in carriers of both variants. Regarding the SYCP2L rs2153157:G>A variant, lower AMH levels were observed in women carrying an AA genotype compared to women carrying a heterozygous genotype (p=0.01). Considering the TDRD3 rs4886238:G>A variant, women carrying an AA genotype presented higher AMH levels than carriers of GG and GA genotypes (p=0.025). Nevertheless, no difference was found regarding response to COS or reproductive outcomes. Considering the combined effect of the variants, women carrying the heterozygous genotype of both variants presented statistically increased AMH levels compared to SYCP2L rs2153157 AA genotype carriers and TDRD3 rs4886238 GG genotype carriers (p=0.042). CONCLUSIONS Individually and combined, the SYCP2L rs2153157 and TDRD3 rs4886238 variants have an effect on AMH level.
Collapse
Affiliation(s)
- Iasmim Ribeiro Rosa
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Caio Parente Barbosa
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| | - Caroline Awoki Ferrandez
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Bianca Del Bel Sonoda
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Denise Maria Christofolini
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| | - Camila Martins Trevisan
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS “Civico - Di Cristina -
Benfratelli”, Department of Health Promotion, Mother and Child Care, Internal
Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Carla Peluso
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
| | - Bianca Bianco
- Discipline of Sexual and Reproductive Health and Populational
Genetics, Department of Collective Health, Faculdade de Medicina do ABC/Centro
Universitário Saúde ABC, FMABC, Santo André, São Paulo,
Brazil
- Instituto Ideia Fértil, Santo André, Brazil
| |
Collapse
|
2
|
Single-Cell RNA Sequencing of the Testis of Ciona intestinalis Reveals the Dynamic Transcriptional Profile of Spermatogenesis in Protochordates. Cells 2022; 11:cells11243978. [PMID: 36552742 PMCID: PMC9776925 DOI: 10.3390/cells11243978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is a complex and continuous process of germ-cell differentiation. This complex process is regulated by many factors, of which gene regulation in spermatogenic cells plays a decisive role. Spermatogenesis has been widely studied in vertebrates, but little is known about spermatogenesis in protochordates. Here, for the first time, we performed single-cell RNA sequencing (scRNA-seq) on 6832 germ cells from the testis of adult Ciona intestinalis. We identified six germ cell populations and revealed dynamic gene expression as well as transcriptional regulation during spermatogenesis. In particular, we identified four spermatocyte subtypes and key genes involved in meiosis in C. intestinalis. There were remarkable similarities and differences in gene expression during spermatogenesis between C. intestinalis and two other vertebrates (Chinese tongue sole and human). We identified many spermatogenic-cell-specific genes with functions that need to be verified. These findings will help to further improve research on spermatogenesis in chordates.
Collapse
|
3
|
Wellard SR, Skinner MW, Zhao X, Shults C, Jordan PW. PLK1 depletion alters homologous recombination and synaptonemal complex disassembly events during mammalian spermatogenesis. Mol Biol Cell 2022; 33:ar37. [PMID: 35274968 PMCID: PMC9282006 DOI: 10.1091/mbc.e21-03-0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 02/25/2022] [Accepted: 03/03/2022] [Indexed: 11/11/2022] Open
Abstract
Homologous recombination (HR) is an essential meiotic process that contributes to the genetic variation of offspring and ensures accurate chromosome segregation. Recombination is facilitated by the formation and repair of programmed DNA double-strand breaks. These DNA breaks are repaired via recombination between maternal and paternal homologous chromosomes and a subset result in the formation of crossovers. HR and crossover formation is facilitated by synapsis of homologous chromosomes by a proteinaceous scaffold structure known as the synaptonemal complex (SC). Recent studies in yeast and worms have indicated that polo-like kinases (PLKs) regulate several events during meiosis, including DNA recombination and SC dynamics. Mammals express four active PLKs (PLK1-4), and our previous work assessing localization and kinase function in mouse spermatocytes suggested that PLK1 coordinates nuclear events during meiotic prophase. Therefore, we conditionally mutated Plk1 in early prophase spermatocytes and assessed stages of HR, crossover formation, and SC processes. Plk1 mutation resulted in increased RPA foci and reduced RAD51/DMC1 foci during zygonema, and an increase of both class I and class II crossover events. Furthermore, the disassembly of SC lateral elements was aberrant. Our results highlight the importance of PLK1 in regulating HR and SC disassembly during spermatogenesis.
Collapse
Affiliation(s)
- Stephen R. Wellard
- Biochemistry and Molecular Biology Department, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Marnie W. Skinner
- Biochemistry and Molecular Biology Department, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Xueqi Zhao
- Biochemistry and Molecular Biology Department, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Chris Shults
- Biochemistry and Molecular Biology Department, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Philip W. Jordan
- Biochemistry and Molecular Biology Department, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| |
Collapse
|
4
|
Hosoya N, Miyagawa K. Synaptonemal complex proteins modulate the level of genome integrity in cancers. Cancer Sci 2021; 112:989-996. [PMID: 33382503 PMCID: PMC7935773 DOI: 10.1111/cas.14791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 01/09/2023] Open
Abstract
The synaptonemal complex (SC) is a proteinaceous structure that is transiently formed during meiosis to promote homologous recombination between maternal and paternal chromosomes. As this structure is required only for meiotic recombination, the proteins constituting the complex are almost undetectable in normal somatic cells, but they can be expressed under the conditions in which the transcriptional machinery is deregulated. Accumulating evidence indicates that they are epigenetically expressed in cancers of various origin. Not surprisingly, in contrast to their meiotic roles, the somatic roles of the SC proteins remain to be investigated. However, it has recently been reported that SYCP3 and SYCE2 control DNA double‐strand break repair negatively and positively, respectively, suggesting that the ectopic expression of the SC proteins in somatic cells could be associated with the maintenance of genomic instability. Thus, it is highly likely that the investigation of the somatic roles of the SC proteins would improve our understanding of the mechanisms underlying tumor development.
Collapse
Affiliation(s)
- Noriko Hosoya
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Ortiz R, Chavero SJ, Echeverría OM, Hernandez-Hernandez A. Synaptonemal complex formation produces a particular arrangement of the lateral element-associated DNA. Exp Cell Res 2021; 399:112455. [PMID: 33400935 DOI: 10.1016/j.yexcr.2020.112455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 11/30/2020] [Accepted: 12/19/2020] [Indexed: 11/27/2022]
Abstract
During meiosis, homologous chromosomes exchange genetic material. This exchange or meiotic recombination is mediated by a proteinaceous scaffold known as the Synaptonemal complex (SC). Any defects in its formation produce failures in meiotic recombination, chromosome segregation and meiosis completion. It has been proposed that DNA repair events that will be resolved by crossover between homologous chromosomes are predetermined by the SC. Hence, structural analysis of the organization of the DNA in the SC could shed light on the process of crossover interference. In this work, we employed an ultrastructural DNA staining technique on mouse testis and followed nuclei of pachytene cells. We observed structures organized similarly to the SCs stained with conventional techniques. These structures, presumably the DNA in the SCs, are delineating the edges of both lateral elements and no staining was observed between them. DNA in the LEs resembles two parallel tracks. However, a bubble-like staining pattern in certain regions of the SC was observed. Furthermore, this staining pattern is found in SCs formed between non-homologous chromosomes, in SCs formed between sister chromatids and in SCs without lateral elements, suggesting that this particular organization of the DNA is determined by the synapsis of the chromosomes despite their lack of homology or the presence of partially formed SCs.
Collapse
Affiliation(s)
- Rosario Ortiz
- Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Silvia Juárez Chavero
- Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Olga M Echeverría
- Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abrahan Hernandez-Hernandez
- Biología de Células Individuales (BIOCELIN), Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, CDMX, Mexico.
| |
Collapse
|
6
|
Ovarian transcriptome analysis of Mactra chinensis provides insights into genes expressed during the intermediate and ripening stages. Anim Reprod Sci 2019; 208:106078. [DOI: 10.1016/j.anireprosci.2019.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/20/2019] [Accepted: 05/10/2019] [Indexed: 11/30/2022]
|
7
|
Sciurano RB, Pigozzi MI, Benavente R. Disassembly of the synaptonemal complex in chicken oocytes analyzed by super-resolution microscopy. Chromosoma 2019; 128:443-451. [PMID: 30793238 DOI: 10.1007/s00412-019-00693-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/11/2019] [Accepted: 02/11/2019] [Indexed: 12/27/2022]
Abstract
The synaptonemal complex is an evolutionarily conserved, supramolecular structure that holds the homologous chromosomes together during the pachytene stage of the first meiotic prophase. Among vertebrates, synaptonemal complex dynamics has been analyzed in mouse spermatocytes following the assembly of its components from leptotene to pachytene stages. With few exceptions, a detailed study of the disassembly of SCs and the behavior of SC components at recombination sites at the onset of diplotene has not been accomplished. Here, we describe for the first time the progressive disassembly of the SC in chicken oocytes during the initial steps of desynapsis using immunolocalization of specific SC proteins and super-resolution microscopy. We found that transverse filament protein SYCP1 and central element component SYCE3 remain associated with the lateral elements at the beginning of chromosomal axis separation. As the separation between lateral elements widens, these proteins eventually disappear, without any evidence of subsequent association. Our observations support the idea that post-translational modifications of the central region components have a role at the initial phases of the SC disassembly. At the crossover sites, signaled by persistent MLH1 foci, the central region proteins are no longer detected when the SYCP3-positive lateral elements are widely separated. These findings are indicative that SC disassembly follows a general pattern along the desynaptic bivalents. The present work shows that the use of avian oocytes at prophase I provides a valuable model to explore the time course and chromosomal localization of SC proteins and its relationship with local changes along meiotic bivalents.
Collapse
Affiliation(s)
- Roberta B Sciurano
- 2da. U.A. Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- CONICET, Buenos Aires, Argentina
| | - María Inés Pigozzi
- Instituto de Investigaciones Biomédicas, INBIOMED, CONICET-Universidad de Buenos Aires, Paraguay 2155, piso 10, C1121ABG, Buenos Aires, Argentina.
| | - Ricardo Benavente
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany.
| |
Collapse
|
8
|
Mary N, Villagómez DAF, Revay T, Rezaei S, Donaldson B, Pinton A, King WA. Meiotic Synapsis and Gene Expression Altered by a Balanced Y-Autosome Reciprocal Translocation in an Azoospermic Pig. Sex Dev 2018; 12:256-263. [PMID: 30179878 DOI: 10.1159/000491804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2018] [Indexed: 12/16/2022] Open
Abstract
Meiotic sex chromosome silencing (MSCS) has been argued as a prerequisite for normal meiotic cell division progression during the synaptic prophase I stage. Furthermore, irregular asynapsis of autosomal axes at meiosis may be encompassing the lack of transcriptional activity normally observed for the X and Y sex chromosomes. Therefore, any chromosomal rearrangement compromising the normal mechanism of MSCS and/or the contrary, the normal meiotic transcriptional activity of autosomal chromosomes, may be observed as a meiotic and concomitant spermatogenesis arrest. Previously, we have described a Y-autosome translocation t(Y;13)(p1.3;q3.3) in an azoospermic boar. Its chromosome synapsis behavior by synaptonemal complex immunostaining and FISH analyses is documented here. Histone γH2AX protein foci appeared to be located at unsynapsed chromosomal segments (e.g., X chromosome univalents or unpaired multivalent segments), although interestingly a high proportion of primary spermatocytes showed full paired synaptonemal complex-multivalent configurations which were devoid of a γH2AX focus signal, indicating meiotic chromosome silencing. RT-qPCR analysis of testicular expression showed downregulation of 3 SSC13 genes (MLH1, SOX2, UBE2B) and upregulation of SSCY genes (ZFY, SRY). The irregularity of the normal transcription pattern in case of these genes with proven roles in the testis is in agreement with the cytological observations and could contribute to the observed phenotype.
Collapse
|
9
|
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: 2.8] [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
|
10
|
Li JY, Pan LQ, Miao JJ, Xu RY, Xu WJ. De novo assembly and characterization of the ovarian transcriptome reveal mechanisms of the final maturation stage in Chinese scallop Chlamys farreri. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2016; 20:118-124. [DOI: 10.1016/j.cbd.2016.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022]
|
11
|
Yan Z, Fan D, Meng Q, Yang J, Zhao W, Guo F, Song D, Guo R, Sun K, Wang J. Transcription factor ZFP38 is essential for meiosis prophase I in male mice. Reproduction 2016; 152:431-7. [PMID: 27492080 DOI: 10.1530/rep-16-0225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/04/2016] [Indexed: 11/08/2022]
Abstract
The production of haploid gametes by meiosis is a cornerstone of sexual reproduction and maintenance of genome integrity. Zfp38 mRNA is expressed in spermatocytes, indicating that transcription factor ZFP38 has the potential to regulate transcription during meiosis. In this study, we generated Zfp38 conditional knockout mice (Zfp38(flox/flox), Stra8-Cre, hereafter called Zfp38 cKO) and found that spermatogenesis did not progress beyond meiosis prophase I in Zfp38 cKO mice. Using a chromosomal spread technique, we observed that Zfp38 cKO spermatocytes exhibited a failure in chromosomal synapsis observed by SYCP1/SYCP3 double staining. Progression of DNA double-strand breaks (DSB) repair is disrupted in Zfp38 cKO spermatocytes, as revealed by γ-H2AX, RAD51 and MLH1 staining. Furthermore, the mRNA and protein levels of DSB repair enzymes and factors that guide their loading onto sites of DSBs, such as RAD51, DMC1, RAD51, TEX15 and PALB2, were significantly reduced in Zfp38 cKO spermatocytes. Taken together, our data suggest that ZFP38 is critical for the chromosomal synapsis and DSB repairs partially via its regulation of DSB repair-associated protein expression during meiotic progression in mouse.
Collapse
Affiliation(s)
- Zechen Yan
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical ScienceZhengzhou, Henan, China
| | - Qingjun Meng
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinjian Yang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Zhao
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fei Guo
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongjian Song
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruiming Guo
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ke Sun
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaxiang Wang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Hemmer LW, Blumenstiel JP. Holding it together: rapid evolution and positive selection in the synaptonemal complex of Drosophila. BMC Evol Biol 2016; 16:91. [PMID: 27150275 PMCID: PMC4857336 DOI: 10.1186/s12862-016-0670-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022] Open
Abstract
Background The synaptonemal complex (SC) is a highly conserved meiotic structure that functions to pair homologs and facilitate meiotic recombination in most eukaryotes. Five Drosophila SC proteins have been identified and localized within the complex: C(3)G, C(2)M, CONA, ORD, and the newly identified Corolla. The SC is required for meiotic recombination in Drosophila and absence of these proteins leads to reduced crossing over and chromosomal nondisjunction. Despite the conserved nature of the SC and the key role that these five proteins have in meiosis in D. melanogaster, they display little apparent sequence conservation outside the genus. To identify factors that explain this lack of apparent conservation, we performed a molecular evolutionary analysis of these genes across the Drosophila genus. Results For the five SC components, gene sequence similarity declines rapidly with increasing phylogenetic distance and only ORD and C(2)M are identifiable outside of the Drosophila genus. SC gene sequences have a higher dN/dS (ω) rate ratio than the genome wide average and this can in part be explained by the action of positive selection in almost every SC component. Across the genus, there is significant variation in ω for each protein. It further appears that ω estimates for the five SC components are in accordance with their physical position within the SC. Components interacting with chromatin evolve slowest and components comprising the central elements evolve the most rapidly. Finally, using population genetic approaches, we demonstrate that positive selection on SC components is ongoing. Conclusions SC components within Drosophila show little apparent sequence homology to those identified in other model organisms due to their rapid evolution. We propose that the Drosophila SC is evolving rapidly due to two combined effects. First, we propose that a high rate of evolution can be partly explained by low purifying selection on protein components whose function is to simply hold chromosomes together. We also propose that positive selection in the SC is driven by its sex-specificity combined with its role in facilitating both recombination and centromere clustering in the face of recurrent bouts of drive in female meiosis. Electronic supplementary material The online version of this article (doi:10.1186/s12862-016-0670-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lucas W Hemmer
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, 66045, USA.
| | - Justin P Blumenstiel
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, 66045, USA
| |
Collapse
|
13
|
Hernández-Hernández A, Masich S, Fukuda T, Kouznetsova A, Sandin S, Daneholt B, Höög C. The central element of the synaptonemal complex in mice is organized as a bilayered junction structure. J Cell Sci 2016; 129:2239-49. [PMID: 27103161 DOI: 10.1242/jcs.182477] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/14/2016] [Indexed: 01/25/2023] Open
Abstract
The synaptonemal complex transiently stabilizes pairing interactions between homologous chromosomes during meiosis. Assembly of the synaptonemal complex is mediated through integration of opposing transverse filaments into a central element, a process that is poorly understood. We have, here, analyzed the localization of the transverse filament protein SYCP1 and the central element proteins SYCE1, SYCE2 and SYCE3 within the central region of the synaptonemal complex in mouse spermatocytes using immunoelectron microscopy. Distribution of immuno-gold particles in a lateral view of the synaptonemal complex, supported by protein interaction data, suggest that the N-terminal region of SYCP1 and SYCE3 form a joint bilayered central structure, and that SYCE1 and SYCE2 localize in between the two layers. We find that disruption of SYCE2 and TEX12 (a fourth central element protein) localization to the central element abolishes central alignment of the N-terminal region of SYCP1. Thus, our results show that all four central element proteins, in an interdependent manner, contribute to stabilization of opposing N-terminal regions of SYCP1, forming a bilayered transverse-filament-central-element junction structure that promotes synaptonemal complex formation and synapsis.
Collapse
Affiliation(s)
| | - Sergej Masich
- Department of Cell and Molecular Biology, Karolinska Institutet Berzelius väg 35, Stockholm 171 77, Sweden
| | - Tomoyuki Fukuda
- Department of Cell and Molecular Biology, Karolinska Institutet Berzelius väg 35, Stockholm 171 77, Sweden Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Anna Kouznetsova
- Department of Cell and Molecular Biology, Karolinska Institutet Berzelius väg 35, Stockholm 171 77, Sweden
| | - Sara Sandin
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | - Bertil Daneholt
- Department of Cell and Molecular Biology, Karolinska Institutet Berzelius väg 35, Stockholm 171 77, Sweden
| | - Christer Höög
- Department of Cell and Molecular Biology, Karolinska Institutet Berzelius väg 35, Stockholm 171 77, Sweden
| |
Collapse
|
14
|
Fraune J, Brochier-Armanet C, Alsheimer M, Volff JN, Schücker K, Benavente R. Evolutionary history of the mammalian synaptonemal complex. Chromosoma 2016; 125:355-60. [PMID: 26968413 DOI: 10.1007/s00412-016-0583-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 12/29/2022]
Abstract
The synaptonemal complex (SC), a key structure of meiosis that assembles during prophase I, has been initially described 60 years ago. Since then, the structure has been described in many sexually reproducing organisms. However, the SC protein components were characterized in only few model organisms. Surprisingly, they lacked an apparent evolutionary relationship despite the conserved structural organization of the SC. For better understanding of this obvious discrepancy, the evolutionary history of the SC and its individual components has been investigated in Metazoa in detail. The results are consistent with the notion of a single origin of the metazoan SC and provide evidence for a dynamic evolutionary history of the SC components. In this mini review, we recapitulate and discuss new insights into metazoan SC evolution.
Collapse
Affiliation(s)
- Johanna Fraune
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Céline Brochier-Armanet
- Université Lyon 1, CNRS, UMR5558, Laboratoire de Biométrie et Biologie Evolutive, 43 bd du 11 novembre 1918, Villeurbanne, 69622, France
| | - Manfred Alsheimer
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Jean-Nicolas Volff
- Institut de Génomique Fonctionnelle de Lyon, École Normale Supérieure de Lyon, CNRS, Université Lyon 1, Lyon, France
| | - Katharina Schücker
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Ricardo Benavente
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany.
| |
Collapse
|
15
|
Zhou J, Stein P, Leu NA, Chmátal L, Xue J, Ma J, Huang X, Lampson MA, Schultz RM, Wang PJ. Accelerated reproductive aging in females lacking a novel centromere protein SYCP2L. Hum Mol Genet 2015; 24:6505-14. [PMID: 26362258 PMCID: PMC4614708 DOI: 10.1093/hmg/ddv359] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/07/2015] [Accepted: 09/01/2015] [Indexed: 11/13/2022] Open
Abstract
Menopause results from loss of ovarian function and marks the end of a woman's reproductive life. Alleles of the human SYCP2L locus are associated with age at natural menopause (ANM). SYCP2L is a paralogue of the synaptonemal complex protein SYCP2 and is expressed exclusively in oocytes. Here we report that SYCP2L localizes to centromeres of dictyate stage oocytes, which represent the limited pool of primordial oocytes that are formed perinatally and remain arrested till ovulation. Centromere localization of SYCP2L requires its C-terminal portion, which is missing in truncated variants resulting from low-frequency nonsense mutations identified in humans. Female mice lacking SYCP2L undergo a significantly higher progressive loss of oocytes with age compared with wild-type females and are less fertile. Specifically, the pool of primordial oocytes becomes more rapidly depleted in SYCP2L-deficient than in wild-type females, such that with aging, fewer oocytes undergo maturation in developing follicles. We find that a human SYCP2L intronic single nucleotide polymorphism (SNP) rs2153157, which is associated with ANM, changes the splicing efficiency of U12-type minor introns and may therefore regulate the steady-state amount of SYCP2L transcript. Furthermore, the more efficiently spliced allele of this intronic SNP in SYCP2L is associated with increased ANM. Our results suggest that SYCP2L promotes the survival of primordial oocytes and thus provide functional evidence for its association with ANM in humans.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Paula Stein
- Department of Biology, University of Pennsylvania, 433 South University Avenue, Philadelphia, PA 19104, USA and
| | - N Adrian Leu
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Lukáš Chmátal
- Department of Biology, University of Pennsylvania, 433 South University Avenue, Philadelphia, PA 19104, USA and
| | - Jiangyang Xue
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Jun Ma
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA, Department of Biology, University of Pennsylvania, 433 South University Avenue, Philadelphia, PA 19104, USA and
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Michael A Lampson
- Department of Biology, University of Pennsylvania, 433 South University Avenue, Philadelphia, PA 19104, USA and
| | - Richard M Schultz
- Department of Biology, University of Pennsylvania, 433 South University Avenue, Philadelphia, PA 19104, USA and
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA,
| |
Collapse
|
16
|
Laissue P. Aetiological coding sequence variants in non-syndromic premature ovarian failure: From genetic linkage analysis to next generation sequencing. Mol Cell Endocrinol 2015; 411:243-57. [PMID: 25960166 DOI: 10.1016/j.mce.2015.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/14/2015] [Accepted: 05/04/2015] [Indexed: 01/19/2023]
Abstract
Premature ovarian failure (POF) is a frequent pathology affecting 1-1.5% of women under 40 years old. Despite advances in diagnosing and treating human infertility, POF is still classified as being idiopathic in 50-80% of cases, strongly suggesting a genetic origin for the disease. Different types of autosomal and X-linked genetic anomalies can originate the phenotype in syndromic and non-syndromic POF cases. Particular interest has been focused on research into non-syndromic POF causative coding variants during the past two decades. This has been based on the assumption that amino acid substitutions might modify the intrinsic physicochemical properties of functional proteins, thereby inducing pathological phenotypes. In this case, a restricted number of mutations might originate the disease. However, like other complex pathologies, POF might result from synergistic/compensatory effects caused by several low-to-mildly drastic mutations which have frequently been classified as non-functional SNPs. Indeed, reproductive phenotypes can be considered as quantitative traits resulting from the subtle interaction of many genes. Although numerous sequencing projects have involved candidate genes, only a few coding mutations explaining a low percentage of cases have been described. Such apparent failure to identify aetiological coding sequence variations might have been due to the inherent molecular complexity of mammalian reproduction and to the difficulty of simultaneously analysing large genomic regions by Sanger sequencing. The purpose of this review is to present the molecular and cellular effects caused by non-synonymous mutations which have been formally associated, by functional tests, with the aetiology of hypergonadotropic non-syndromic POF. Considerations have also been included regarding the polygenic nature of reproduction and POF, as well as future approaches for identifying novel aetiological genes based on next generation sequencing (NGS).
Collapse
Affiliation(s)
- Paul Laissue
- Unidad de Genética, Grupo GENIUROS, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
17
|
The T657C polymorphism on the SYCP3 gene is associated with recurrent pregnancy loss. J Assist Reprod Genet 2014; 31:1377-81. [PMID: 25059562 DOI: 10.1007/s10815-014-0272-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/01/2014] [Indexed: 12/31/2022] Open
Abstract
SYCP3 (Sinaptonemal complex protein 3) plays a critical role in pairing and recombination of homologous chromosomes in meiosis 1. It has been shown that lack of this gene leads to infertility in male and weakened fertility in female mice. In a case-control study, we investigated the SYCP3T657C polymorphism in the genome of 100 Iranian women with recurrent pregnancy losses of unknown causes as well as 100 control samples of normal fertile women having at least one healthy child. The general aim of our study was to determine whether there is a relationship between genetic changes in the SYCP3 gene and recurrent pregnancy loss in human or not. Frequency of the heterozygous genotype and mutated allele C were significantly higher in women with recurrent pregnancy losses (P-value < 0.005). Our findings suggest that the T657C polymorphism of the SYCP3 gene is possibly associated with recurrent pregnancy loss of unknown cause in human.
Collapse
|
18
|
Zhuang XJ, Shi YQ, Xu B, Chen L, Tang WH, Huang J, Lian Y, Liu P, Qiao J. SLX2 interacting with BLOS2 is differentially expressed during mouse oocyte meiotic maturation. Cell Cycle 2014; 13:2231-7. [PMID: 24870619 DOI: 10.4161/cc.29265] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gametogenesis is a complex biological process of producing cells for sexual reproduction. Xlr super family members containing a conserved COR1 domain play essential roles in gametogenesis. In the present study, we identified that Slx2, a novel member of Xlr super family, is specifically expressed in the meiotic oocytes, which is demonstrated by western blotting and immunohistochemistry studies. In the first meiotic prophase, SLX2 is unevenly distributed in the nuclei of oocytes, during which phase SLX2 is partly co-localized with SYCP3 in synaptonemal complex and γH2AX in the nucleus of oocytes. Interestingly, the localization of SLX2 was found to be switched into the cytoplasm of oocytes after prometaphase I during oocyte maturation. Furthermore, yeast two-hybrid and coimmunoprecipitation studies demonstrated that SLX2 interacts with BLOS2, which is a novel centrosome-associated protein, and co-localized with γ-Tubulin, which is a protein marker of chromosome segregation in meiosis. These results indicated that SLX2 might get involved in chromosomes segregation during meiosis by interaction with BLOS2. In conclusion, SLX2 might be a novel gametogenesis-related protein that could play multiple roles in regulation of meiotic processes including synaptonemal complex assembly and chromosome segregation.
Collapse
Affiliation(s)
- Xin-Jie Zhuang
- Center for Reproductive Medicine; Department of Obstetrics and Gynecology; Key Laboratory of Assisted Reproduction; Ministry of Education; Peking University Third Hospital; Beijing, PR China
| | - Yu-Qiang Shi
- College of Bio-Engineering; Weifang University; Weifang, PR China
| | - Bo Xu
- Center for Reproductive Medicine; Anhui Provincial Hospital Affiliated to Anhui Medical University; Hefei, PR China
| | - Lei Chen
- Reproductive Medical Centre; First Affiliated Hospital of Zhengzhou University; Zhengzhou, PR China
| | - Wen-Hao Tang
- Department of Urology; the Third Hospital of Peking University; Beijing, PR China
| | - Jin Huang
- Center for Reproductive Medicine; Department of Obstetrics and Gynecology; Key Laboratory of Assisted Reproduction; Ministry of Education; Peking University Third Hospital; Beijing, PR China
| | - Ying Lian
- Center for Reproductive Medicine; Department of Obstetrics and Gynecology; Key Laboratory of Assisted Reproduction; Ministry of Education; Peking University Third Hospital; Beijing, PR China
| | - Ping Liu
- Center for Reproductive Medicine; Department of Obstetrics and Gynecology; Key Laboratory of Assisted Reproduction; Ministry of Education; Peking University Third Hospital; Beijing, PR China
| | - Jie Qiao
- Center for Reproductive Medicine; Department of Obstetrics and Gynecology; Key Laboratory of Assisted Reproduction; Ministry of Education; Peking University Third Hospital; Beijing, PR China
| |
Collapse
|
19
|
Jiang H, Wang L, Cui Y, Xu Z, Guo T, Cheng D, Xu P, Yu W, Shi Q. Meiotic Chromosome Behavior in a Human Male t(8;15) Carrier. J Genet Genomics 2014; 41:177-85. [DOI: 10.1016/j.jgg.2014.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
|
20
|
Biswas U, Wetzker C, Lange J, Christodoulou EG, Seifert M, Beyer A, Jessberger R. Meiotic cohesin SMC1β provides prophase I centromeric cohesion and is required for multiple synapsis-associated functions. PLoS Genet 2013; 9:e1003985. [PMID: 24385917 PMCID: PMC3873225 DOI: 10.1371/journal.pgen.1003985] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 10/14/2013] [Indexed: 01/09/2023] Open
Abstract
Cohesin subunit SMC1β is specific and essential for meiosis. Previous studies showed functions of SMC1β in determining the axis-loop structure of synaptonemal complexes (SCs), in providing sister chromatid cohesion (SCC) in metaphase I and thereafter, in protecting telomere structure, and in synapsis. However, several central questions remained unanswered and concern roles of SMC1β in SCC and synapsis and processes related to these two processes. Here we show that SMC1β substantially supports prophase I SCC at centromeres but not along chromosome arms. Arm cohesion and some of centromeric cohesion in prophase I are provided by non-phosphorylated SMC1α. Besides supporting synapsis of autosomes, SMC1β is also required for synapsis and silencing of sex chromosomes. In absence of SMC1β, the silencing factor γH2AX remains associated with asynapsed autosomes and fails to localize to sex chromosomes. Microarray expression studies revealed up-regulated sex chromosome genes and many down-regulated autosomal genes. SMC1β is further required for non-homologous chromosome associations observed in absence of SPO11 and thus of programmed double-strand breaks. These breaks are properly generated in Smc1β−/− spermatocytes, but their repair is delayed on asynapsed chromosomes. SMC1α alone cannot support non-homologous associations. Together with previous knowledge, three main functions of SMC1β have emerged, which have multiple consequences for spermatocyte biology: generation of the loop-axis architecture of SCs, homologous and non-homologous synapsis, and SCC starting in early prophase I. The generation of mammalian gametes through meiosis comprises two subsequent cell divisions. The first division, meiosis I, features highly specific chromosome structures, and behavior, and requires distinct sets of chromosome-associated proteins. Cohesin proteins, of which some are meiosis-specific, are essential for meiosis, but their particular roles in meiosis are incompletely understood. We show here that SMC1β, a meiosis-specific cohesin, serves key functions already in prophase of meiosis I: SMC1β contributes to keeping sister chromatids in cohesion at their centromeres and supports synapsis of the four sister chromatids present in these cells. SMC1β is required for the synapsis of the X and Y sex chromosomes. The failure of autosomes to properly synapse in absence of SMC1β causes extensive alterations in gene expression. This leads to expression of sex chromosome-linked genes, which are lethal at this stage, explaining the death of spermatocytes in mid-prophase I. Together with the analyses of other cohesin proteins and of phosphorylated forms of SMC3 and SMC1α, this paper describes hitherto undescribed properties and functions of meiotic cohesin in sister chromatid cohesion and synapsis.
Collapse
Affiliation(s)
- Uddipta Biswas
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Cornelia Wetzker
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Julian Lange
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | | | - Andreas Beyer
- Biotechnology Center, TU Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Rolf Jessberger
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
21
|
Human disease locus discovery and mapping to molecular pathways through phylogenetic profiling. Mol Syst Biol 2013; 9:692. [PMID: 24084807 PMCID: PMC3817400 DOI: 10.1038/msb.2013.50] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/29/2013] [Indexed: 12/16/2022] Open
Abstract
By analyzing the conservation of human proteins across 87 species, we sorted proteins into clusters of coevolution. Some clusters are enriched for genes assigned to particular human diseases or molecular pathways; the other genes in the same cluster may function in related pathways and diseases. ![]()
Many genes that were thought to map to different diseases are actually coevolved together and mapped into the same phylogenetic clusters. Many molecular pathways map to the same phylogenetic clusters as genes associated with specific human diseases. Focusing on proteins coevolved with the microphthalmia-associated transcription factor (MITF), we identified the Notch pathway suppressor of hairless (RBP-Jk/SuH) transcription factor, and showed that RBP-Jk functions as an MITF cofactor. Our analysis thus establishes a connectivity between different diseases and pathways, linking diseases phenotypes and functional gene groups.
Genes with common profiles of the presence and absence in disparate genomes tend to function in the same pathway. By mapping all human genes into about 1000 clusters of genes with similar patterns of conservation across eukaryotic phylogeny, we determined that sets of genes associated with particular diseases have similar phylogenetic profiles. By focusing on those human phylogenetic gene clusters that significantly overlap some of the thousands of human gene sets defined by their coexpression or annotation to pathways or other molecular attributes, we reveal the evolutionary map that connects molecular pathways and human diseases. The other genes in the phylogenetic clusters enriched for particular known disease genes or molecular pathways identify candidate genes for roles in those same disorders and pathways. Focusing on proteins coevolved with the microphthalmia-associated transcription factor (MITF), we identified the Notch pathway suppressor of hairless (RBP-Jk/SuH) transcription factor, and showed that RBP-Jk functions as an MITF cofactor.
Collapse
|
22
|
Phylogenies of central element proteins reveal the dynamic evolutionary history of the mammalian synaptonemal complex: ancient and recent components. Genetics 2013; 195:781-93. [PMID: 24026100 DOI: 10.1534/genetics.113.156679] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During meiosis, the stable pairing of the homologous chromosomes is mediated by the assembly of the synaptonemal complex (SC). Its tripartite structure is well conserved in Metazoa and consists of two lateral elements (LEs) and a central region (CR) that in turn is formed by several transverse filaments (TFs) and a central element (CE). In a previous article, we have shown that not only the structure, but also the major structural proteins SYCP1 (TFs) and SYCP3 (LEs) of the mammalian SC are conserved in metazoan evolution. In continuation of this work, we now investigated the evolution of the mammalian CE-specific proteins using phylogenetic and biochemical/cytological approaches. In analogy to the observations made for SYCP1 and SYCP3, we did not detect homologs of the mammalian CE proteins in insects or nematodes, but in several other metazoan clades. We were able to identify homologs of three mammalian CE proteins in several vertebrate and invertebrate species, for two of these proteins down to the basal-branching phylum of Cnidaria. Our approaches indicate that the SC arose only once, but evolved dynamically during diversification of Metazoa. Certain proteins appear to be ancient in animals, but successive addition of further components as well as protein loss and/or replacements have also taken place in some lineages.
Collapse
|
23
|
Shi YQ, Zhuang XJ, Xu B, Hua J, Liao SY, Shi Q, Cooke HJ, Han C. SYCP3-like X-linked 2 is expressed in meiotic germ cells and interacts with synaptonemal complex central element protein 2 and histone acetyltransferase TIP60. Gene 2013; 527:352-9. [DOI: 10.1016/j.gene.2013.06.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/21/2013] [Accepted: 06/04/2013] [Indexed: 12/28/2022]
|
24
|
Analysis of SYCP3 encoding synaptonemal complex protein 3 in human aneuploidies. Arch Gynecol Obstet 2013; 288:1153-8. [PMID: 23677416 DOI: 10.1007/s00404-013-2861-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/16/2013] [Indexed: 01/14/2023]
|
25
|
Gan H, Cai T, Lin X, Wu Y, Wang X, Yang F, Han C. Integrative proteomic and transcriptomic analyses reveal multiple post-transcriptional regulatory mechanisms of mouse spermatogenesis. Mol Cell Proteomics 2013; 12:1144-57. [PMID: 23325766 DOI: 10.1074/mcp.m112.020123] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian spermatogenesis consists of many cell types and biological processes and serves as an excellent model for studying gene regulation at transcriptional and post-transcriptional levels. Many key proteins, miRNAs, and perhaps piRNAs have been shown to be involved in post-transcriptional regulation of spermatogenesis. However, a systematic method for assessing the relationship between protein and mRNA expression has not been available for studying mechanisms of post-transcriptional regulation. In the present study, we used the iTRAQ-based quantitative proteomic approach to identify 2008 proteins in mouse type A spermatogonia, pachytene spermatocytes, round spermatids, and elongative spermatids with high confidence. Of these proteins, 1194 made up four dynamically changing clusters, which reflect the mitotic amplification, meiosis, and post-meiotic development of germ cells. We identified five major regulatory mechanisms termed "transcript only," "transcript degradation," "translation repression," "translation de-repression," and "protein degradation" based on changes in protein level relative to changes in mRNA level at the mitosis/meiosis transition and the meiosis/post-meiotic development transition. We found that post-transcriptional regulatory mechanisms are related to the generation of piRNAs and antisense transcripts. Our results provide a valuable inventory of proteins produced during mouse spermatogenesis and contribute to elucidating the mechanisms of the post-transcriptional regulation of gene expression in mammalian spermatogenesis.
Collapse
Affiliation(s)
- Haiyun Gan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, PR China
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Sister chromatid cohesion depends on cohesin, a tripartite complex that forms ring structures to hold sister chromatids together in mitosis and meiosis. Meiocytes feature a multiplicity of distinct cohesin proteins and complexes, some meiosis specific, which serve additional functions such as supporting synapsis of two pairs of sister chromatids and determining the loop-axis architecture of prophase I chromosomes. Despite considerable new insights gained in the past few years into the localization and function of some cohesin proteins, and the recent identification of yet another meiosis-specific cohesin subunit, a plethora of open questions remains, which concern not only fundamental germ cell biology but also the consequences of cohesin impairment for human reproductive health.
Collapse
Affiliation(s)
- François McNicoll
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | | | | |
Collapse
|
27
|
de Vries M, Ramos L, de Boer P. Immunofluorescent characterization of meiotic recombination in human males with variable spermatogenesis. Andrology 2012; 1:262-73. [DOI: 10.1111/j.2047-2927.2012.00039.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 11/30/2022]
Affiliation(s)
- M. de Vries
- Department of Obstetrics and Gynaecology; Radboud University Nijmegen Medical Centre; Nijmegen; 6500 HB; The Netherlands
| | - L. Ramos
- Department of Obstetrics and Gynaecology; Radboud University Nijmegen Medical Centre; Nijmegen; 6500 HB; The Netherlands
| | - P. de Boer
- Department of Obstetrics and Gynaecology; Radboud University Nijmegen Medical Centre; Nijmegen; 6500 HB; The Netherlands
| |
Collapse
|
28
|
Jordan PW, Karppinen J, Handel MA. Polo-like kinase is required for synaptonemal complex disassembly and phosphorylation in mouse spermatocytes. J Cell Sci 2012; 125:5061-72. [PMID: 22854038 PMCID: PMC3533391 DOI: 10.1242/jcs.105015] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2012] [Indexed: 01/05/2023] Open
Abstract
During meiosis, accurate coordination of the completion of homologous recombination and synaptonemal complex (SC) disassembly during the prophase to metaphase I (G2/MI) transition is essential to avoid aneuploid gametes and infertility. Previous studies have shown that kinase activity is required to promote meiotic prophase exit. The first step of the G2/MI transition is the disassembly of the central element components of the SC; however, the kinase(s) required to trigger this process remains unknown. Here we assess roles of polo-like kinases (PLKs) in mouse spermatocytes, both in vivo and during prophase exit induced ex vivo by the phosphatase inhibitor okadaic acid. All four PLKs are expressed during the first wave of spermatogenesis. Only PLK1 (not PLK2-4) localizes to the SC during the G2/MI transition. The SC central element proteins SYCP1, TEX12 and SYCE1 are phosphorylated during the G2/MI transition. However, treatment of pachytene spermatocytes with the PLK inhibitor BI 2536 prevented the okadaic-acid-induced meiotic prophase exit and inhibited phosphorylation of the central element proteins as well as their removal from the SC. Phosphorylation assays in vitro demonstrated that PLK1, but not PLK2-4, phosphorylates central element proteins SYCP1 and TEX12. These findings provide mechanistic details of the first stage of SC disassembly in mammalian spermatocytes, and reveal that PLK-mediated phosphorylation of central element proteins is required for meiotic prophase exit.
Collapse
Affiliation(s)
| | - Jesse Karppinen
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- College of the Atlantic, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
29
|
Hydra meiosis reveals unexpected conservation of structural synaptonemal complex proteins across metazoans. Proc Natl Acad Sci U S A 2012; 109:16588-93. [PMID: 23012415 DOI: 10.1073/pnas.1206875109] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The synaptonemal complex (SC) is a key structure of meiosis, mediating the stable pairing (synapsis) of homologous chromosomes during prophase I. Its remarkable tripartite structure is evolutionarily well conserved and can be found in almost all sexually reproducing organisms. However, comparison of the different SC protein components in the common meiosis model organisms Saccharomyces cerevisiae, Arabidopsis thaliana, Caenorhabditis elegans, Drosophila melanogaster, and Mus musculus revealed no sequence homology. This discrepancy challenged the hypothesis that the SC arose only once in evolution. To pursue this matter we focused on the evolution of SYCP1 and SYCP3, the two major structural SC proteins of mammals. Remarkably, our comparative bioinformatic and expression studies revealed that SYCP1 and SYCP3 are also components of the SC in the basal metazoan Hydra. In contrast to previous assumptions, we therefore conclude that SYCP1 and SYCP3 form monophyletic groups of orthologous proteins across metazoans.
Collapse
|
30
|
Fraune J, Schramm S, Alsheimer M, Benavente R. The mammalian synaptonemal complex: protein components, assembly and role in meiotic recombination. Exp Cell Res 2012; 318:1340-6. [PMID: 22394509 DOI: 10.1016/j.yexcr.2012.02.018] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 10/28/2022]
Abstract
The synaptonemal complex (SC) is a proteinaceous structure of chromosome bivalents whose assembly is indispensable for the successful progression of the first meiotic division of sexually reproducing organisms. In this mini-review we will focus on recent progress dealing with the composition and assembly of the mammalian SC. These advances mainly resulted from the systematic use of knockout mice for all known mammalian SC proteins as well as from protein polymerization studies performed in heterologous systems.
Collapse
Affiliation(s)
- Johanna Fraune
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, D-97074 Würzburg, Germany
| | | | | | | |
Collapse
|
31
|
Davies OR, Maman JD, Pellegrini L. Structural analysis of the human SYCE2-TEX12 complex provides molecular insights into synaptonemal complex assembly. Open Biol 2012; 2:120099. [PMID: 22870393 PMCID: PMC3411106 DOI: 10.1098/rsob.120099] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/26/2012] [Indexed: 11/12/2022] Open
Abstract
The successful completion of meiosis is essential for all sexually reproducing organisms. The synaptonemal complex (SC) is a large proteinaceous structure that holds together homologous chromosomes during meiosis, providing the structural framework for meiotic recombination and crossover formation. Errors in SC formation are associated with infertility, recurrent miscarriage and aneuploidy. The current lack of molecular information about the dynamic process of SC assembly severely restricts our understanding of its function in meiosis. Here, we provide the first biochemical and structural analysis of an SC protein component and propose a structural basis for its function in SC assembly. We show that human SC proteins SYCE2 and TEX12 form a highly stable, constitutive complex, and define the regions responsible for their homotypic and heterotypic interactions. Biophysical analysis reveals that the SYCE2-TEX12 complex is an equimolar hetero-octamer, formed from the association of an SYCE2 tetramer and two TEX12 dimers. Electron microscopy shows that biochemically reconstituted SYCE2-TEX12 complexes assemble spontaneously into filamentous structures that resemble the known physical features of the SC central element (CE). Our findings can be combined with existing biological data in a model of chromosome synapsis driven by growth of SYCE2-TEX12 higher-order structures within the CE of the SC.
Collapse
Affiliation(s)
- Owen R Davies
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Old Addenbrookes Site, Cambridge CB2 1GA, UK.
| | | | | |
Collapse
|
32
|
Qiao H, Chen JK, Reynolds A, Höög C, Paddy M, Hunter N. Interplay between synaptonemal complex, homologous recombination, and centromeres during mammalian meiosis. PLoS Genet 2012; 8:e1002790. [PMID: 22761591 PMCID: PMC3386176 DOI: 10.1371/journal.pgen.1002790] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 05/10/2012] [Indexed: 11/24/2022] Open
Abstract
The intimate synapsis of homologous chromosome pairs (homologs) by synaptonemal complexes (SCs) is an essential feature of meiosis. In many organisms, synapsis and homologous recombination are interdependent: recombination promotes SC formation and SCs are required for crossing-over. Moreover, several studies indicate that initiation of SC assembly occurs at sites where crossovers will subsequently form. However, recent analyses in budding yeast and fruit fly imply a special role for centromeres in the initiation of SC formation. In addition, in budding yeast, persistent SC–dependent centromere-association facilitates the disjunction of chromosomes that have failed to become connected by crossovers. Here, we examine the interplay between SCs, recombination, and centromeres in a mammal. In mouse spermatocytes, centromeres do not serve as SC initiation sites and are invariably the last regions to synapse. However, centromeres are refractory to de-synapsis during diplonema and remain associated by short SC fragments. Since SC–dependent centromere association is lost before diakinesis, a direct role in homolog segregation seems unlikely. However, post–SC disassembly, we find evidence of inter-centromeric connections that could play a more direct role in promoting homolog biorientation and disjunction. A second class of persistent SC fragments is shown to be crossover-dependent. Super-resolution structured-illumination microscopy (SIM) reveals that these structures initially connect separate homolog axes and progressively diminish as chiasmata form. Thus, DNA crossing-over (which occurs during pachynema) and axis remodeling appear to be temporally distinct aspects of chiasma formation. SIM analysis of the synapsis and crossover-defective mutant Sycp1−/− implies that SCs prevent unregulated fusion of homolog axes. We propose that SC fragments retained during diplonema stabilize nascent bivalents and help orchestrate local chromosome reorganization that promotes centromere and chiasma function. Gamete cells, such as sperm and eggs, form via the specialized cell division called meiosis. Essential and interdependent features of meiosis include the pairing, recombination, and segregation of maternal and paternal chromosomes. Chromosome pairing culminates with formation of synaptonemal complexes (SCs), zipper-like structures that connect the structural cores or axes of homologous chromosomes. Although SC is known to be important for crossover recombination, details of its function remain enigmatic. In this study, we analyze mouse spermatocytes to investigate the interplay between SC, recombination, and centromeres (the structures that direct chromosome segregation). We show that SC prevents unregulated interactions between chromosome axes. This function appears to be especially important at chromosome ends and at crossover sites where DNA exchange must be coordinated with structural exchange of chromosome axes. We also show that centromeres remain associated by short fragments of SC after general chromosome desynapsis has occurred. Furthermore, we detect a distinct type of inter-centromeric connection that persists even after centromeres desynapse. Such connections may facilitate the segregation of chromosomes that have failed to crossover. Together, our data provide new insights into the functions of SC and raise the possibility of a back-up chromosome segregation system in mammals analogous to those described in fruit flies and budding yeast.
Collapse
Affiliation(s)
- Huanyu Qiao
- Howard Hughes Medical Institute and Departments of Microbiology, Molecular and Cellular Biology, and Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
| | - Jefferson K. Chen
- Howard Hughes Medical Institute and Departments of Microbiology, Molecular and Cellular Biology, and Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
| | - April Reynolds
- Howard Hughes Medical Institute and Departments of Microbiology, Molecular and Cellular Biology, and Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
| | - Christer Höög
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Paddy
- Microscopy and Imaging Facility, Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - Neil Hunter
- Howard Hughes Medical Institute and Departments of Microbiology, Molecular and Cellular Biology, and Cell Biology and Human Anatomy, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
La Salle S, Palmer K, O'Brien M, Schimenti JC, Eppig J, Handel MA. Spata22, a novel vertebrate-specific gene, is required for meiotic progress in mouse germ cells. Biol Reprod 2012; 86:45. [PMID: 22011390 DOI: 10.1095/biolreprod.111.095752] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The N-ethyl-N-nitrosourea-induced repro42 mutation, identified by a forward genetics strategy, causes both male and female infertility, with no other apparent phenotypes. Positional cloning led to the discovery of a nonsense mutation in Spata22, a hitherto uncharacterized gene conserved among bony vertebrates. Expression of both transcript and protein is restricted predominantly to germ cells of both sexes. Germ cells of repro42 mutant mice express Spata22 transcript, but not SPATA22 protein. Gametogenesis is profoundly affected by the mutation, and germ cells in repro42 mutant mice do not progress beyond early meiotic prophase, with subsequent germ cell loss in both males and females. The Spata22 gene is essential for one or more key events of early meiotic prophase, as homologous chromosomes of mutant germ cells do not achieve normal synapsis or repair meiotic DNA double-strand breaks. The repro42 mutation thus identifies a novel mammalian germ cell-specific gene required for meiotic progression.
Collapse
|
34
|
Naranjo T. Finding the correct partner: the meiotic courtship. SCIENTIFICA 2012; 2012:509073. [PMID: 24278707 PMCID: PMC3820632 DOI: 10.6064/2012/509073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 07/15/2012] [Indexed: 05/02/2023]
Abstract
Homologous chromosomes are usually separated at the entrance of meiosis; how they become paired is one of the outstanding mysteries of the meiotic process. Reduction of spacing between homologues makes possible the occurrence of chromosomal interactions leading to homology detection and the formation of bivalents. In many organisms, telomere-led chromosome movements are generated that bring homologues together. Additional movements produced by chromatin conformational changes at early meiosis may also facilitate homologous contacts. Organisms used in the study of meiosis show a surprising variety of strategies for homology detection. In dipterans, homologous chromosomes remain paired throughout most of development. Pairing seems to arise as a balance between promoter and suppressor pairing genes. Some fungi, plants and animals, use mechanisms based on recombinational interactions. Other mechanisms leading to homology search are recombination-independent and require specialized pairing sites. In the worm Caenorhabditis elegans, each chromosome carries a pairing center consisting of a chromosome-specific DNA-protein complex, and in the fission yeast Schizosaccharomyces pombe, the sme2 locus encodes a meiosis-specific non-coding RNA that mediates on homologous recognition. In addition, mismatch correction plays a relevant role, especially in polyploids, which evolved genetic systems that suppress pairing between non-homologous related (homoeologus) chromosomes.
Collapse
Affiliation(s)
- Tomás Naranjo
- Departamento de Genética, Facultad de Biología, Universidad Complutense de Madrid, 28040 Madrid, Spain
- *Tomás Naranjo:
| |
Collapse
|
35
|
A novel mouse synaptonemal complex protein is essential for loading of central element proteins, recombination, and fertility. PLoS Genet 2011; 7:e1002088. [PMID: 21637789 PMCID: PMC3102746 DOI: 10.1371/journal.pgen.1002088] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 03/31/2011] [Indexed: 12/29/2022] Open
Abstract
The synaptonemal complex (SC) is a proteinaceous, meiosis-specific structure that is highly conserved in evolution. During meiosis, the SC mediates synapsis of homologous chromosomes. It is essential for proper recombination and segregation of homologous chromosomes, and therefore for genome haploidization. Mutations in human SC genes can cause infertility. In order to gain a better understanding of the process of SC assembly in a model system that would be relevant for humans, we are investigating meiosis in mice. Here, we report on a newly identified component of the murine SC, which we named SYCE3. SYCE3 is strongly conserved among mammals and localizes to the central element (CE) of the SC. By generating a Syce3 knockout mouse, we found that SYCE3 is required for fertility in both sexes. Loss of SYCE3 blocks synapsis initiation and results in meiotic arrest. In the absence of SYCE3, initiation of meiotic recombination appears to be normal, but its progression is severely impaired resulting in complete absence of MLH1 foci, which are presumed markers of crossovers in wild-type meiocytes. In the process of SC assembly, SYCE3 is required downstream of transverse filament protein SYCP1, but upstream of the other previously described CE–specific proteins. We conclude that SYCE3 enables chromosome loading of the other CE–specific proteins, which in turn would promote synapsis between homologous chromosomes. Meiosis is a special type of cell division that takes place in the germ line of sexually reproducing diploid organisms. Major events during meiosis are the pairing, recombination, and segregation of homologous chromosomes. As a consequence, daughter cells are haploid and genetically diverse. Therefore, meiosis is of utmost importance for the life of sexually reproducing species as it maintains the species-specific chromosome number and generates genetic diversity within a species. Proper segregation of homologous chromosomes during meiosis requires homolog pairs to be physically linked. The synaptonemal complex (SC), a meiosis-specific structure conserved in evolution, is essential for this process. Defective assembly of the SC has deleterious effects on germ cells and can cause infertility in mice and humans. Here, we report on a newly identified protein component of the mammalian SC that we have named SYCE3. SYCE3 is strongly conserved among mammals. Using the mouse as a model system, we demonstrate that loss of SYCE3 leads to infertility in both sexes. Infertility is caused by disruption of meiosis due to the inability of Syce3−/− mice to assemble the central element of SCs. Our findings provide new insights into the complexity of SC assembly and its relevance to mammalian fertility.
Collapse
|
36
|
Abstract
What drives defective spermatocytes into apoptosis during mid-pachytene? A recent study identifies the first mid-pachytene 'killer' genes: two Y-linked transcription factors, the Zfy1/2 gene pair, must be silenced to avoid apoptosis.
Collapse
|
37
|
Youds JL, Boulton SJ. The choice in meiosis – defining the factors that influence crossover or non-crossover formation. J Cell Sci 2011; 124:501-13. [DOI: 10.1242/jcs.074427] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Meiotic crossovers are essential for ensuring correct chromosome segregation as well as for creating new combinations of alleles for natural selection to take place. During meiosis, excess meiotic double-strand breaks (DSBs) are generated; a subset of these breaks are repaired to form crossovers, whereas the remainder are repaired as non-crossovers. What determines where meiotic DSBs are created and whether a crossover or non-crossover will be formed at any particular DSB remains largely unclear. Nevertheless, several recent papers have revealed important insights into the factors that control the decision between crossover and non-crossover formation in meiosis, including DNA elements that determine the positioning of meiotic DSBs, and the generation and processing of recombination intermediates. In this review, we focus on the factors that influence DSB positioning, the proteins required for the formation of recombination intermediates and how the processing of these structures generates either a crossover or non-crossover in various organisms. A discussion of crossover interference, assurance and homeostasis, which influence crossing over on a chromosome-wide and genome-wide scale – in addition to current models for the generation of interference – is also included. This Commentary aims to highlight recent advances in our understanding of the factors that promote or prevent meiotic crossing over.
Collapse
Affiliation(s)
- Jillian L. Youds
- DNA Damage Response Laboratory, Cancer Research UK, London Research Institute, Clare Hall, Blanche Lane, South Mimms EN6 3LD, UK
| | - Simon J. Boulton
- DNA Damage Response Laboratory, Cancer Research UK, London Research Institute, Clare Hall, Blanche Lane, South Mimms EN6 3LD, UK
| |
Collapse
|
38
|
Vranis NM, Van der Heijden GW, Malki S, Bortvin A. Synaptonemal complex length variation in wild-type male mice. Genes (Basel) 2010; 1:505-20. [PMID: 24710100 PMCID: PMC3966217 DOI: 10.3390/genes1030505] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 11/18/2010] [Accepted: 12/08/2010] [Indexed: 11/16/2022] Open
Abstract
Meiosis yields haploid gametes following two successive divisions of a germ cell in the absence of intervening DNA replication. Balanced segregation of homologous chromosomes in Meiosis I is aided by a proteinaceous structure, the synaptonemal complex (SC). The objective of this study was to determine total average autosomal SC lengths in spermatocytes in three commonly used mouse strains (129S4/SvJae, C57BL/6J, and BALB/c). Our experiments revealed that the total autosomal SC length in BALB/c spermatocytes is 9% shorter than in the two other strains. Shorter SCs are also observed in spermatocytes of (BALB/c × 129S4/SvJae) and (C57BL/6J × BALB/c) F1 hybrids suggesting a genetic basis of SC length regulation. Along these lines, we studied expression of a selected group of genes implicated in meiotic chromosome architecture. We found that BALB/c testes express up to 6-fold less of Rec8 mRNA and 4-fold less of REC8 protein. These results suggest that the mechanism that defines the SC length operates via a REC8‑dependent process. Finally, our results demonstrate that genetic background can have an effect on meiotic studies in mice.
Collapse
Affiliation(s)
| | | | - Safia Malki
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA.
| | - Alex Bortvin
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA.
| |
Collapse
|
39
|
Abstract
Sister chromatids are held together from the time of their formation in S phase until they segregate in anaphase by the cohesin complex. In meiosis of most organisms, the mitotic Mcd1/Scc1/Rad21 subunit of the cohesin complex is largely replaced by its paralog named Rec8. This article reviews the specialized functions of Rec8 that are crucial for diverse aspects of chromosome dynamics in meiosis, and presents some speculations relating to meiotic chromosome organization.
Collapse
|
40
|
Schulz RW, de França LR, Lareyre JJ, Le Gac F, Chiarini-Garcia H, Nobrega RH, Miura T. Spermatogenesis in fish. Gen Comp Endocrinol 2010; 165:390-411. [PMID: 19348807 DOI: 10.1016/j.ygcen.2009.02.013] [Citation(s) in RCA: 717] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Accepted: 02/20/2009] [Indexed: 02/06/2023]
Abstract
Spermatogenesis is a developmental process during which a small number of diploid spermatogonial stem cells produce a large number of highly differentiated spermatozoa carrying a haploid, recombined genome. We characterise morphologically the different germ cell stages with particular attention for the spermatogonial generations, including the stem cells and their specific capacity to colonise a recipient's testis after transplantation. We propose a nomenclature for fish germ cells to improve the comparability among different teleost fish but also to higher vertebrates. Survival and development of germ cells depends on their continuous and close contact to Sertoli cells, and we review their multiple roles in the cystic mode of spermatogenesis seen in fish. We then discuss gene expression patterns associated with testis maturation. The endocrine system of vertebrates has evolved as master control system over spermatogenesis. In fish, both pituitary gonadotropins LH and FSH stimulate gonadal sex steroid hormone production directly by activating Leydig cells. Information is reviewed on the effects of progestin, androgens, and estrogens on global testicular gene expression patterns (microarray analysis), and on the molecular mechanisms by which steroids regulate specific candidate genes (identified by subtractive hybridization approaches) during early stages of testis maturation. Moreover, progestin and androgen effects on spermiation and milt hydration are discussed. Sex steroids mainly act via receptors expressed by Sertoli cells. One type of response is that Sertoli cells change growth factor expression, which subsequently modulates germ cell proliferation/differentiation via mechanisms yet to be characterised. Finally, we review data on germ cell autonomous processes, mainly derived from loss-of-function mutant fish lines, before identifying a number of focus areas for future research activities.
Collapse
Affiliation(s)
- Rüdiger W Schulz
- Utrecht University, Science Faculty, Department Biology, Padualaan 8, NL-3584 CH Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Meiosis is an essential stage in gamete formation in all sexually reproducing organisms. Studies of mutations in model organisms and of human haplotype patterns are leading to a clearer understanding of how meiosis has adapted from yeast to humans, the genes that control the dynamics of chromosomes during meiosis, and how meiosis is tied to gametic success. Genetic disruptions and meiotic errors have important roles in infertility and the aetiology of developmental defects, especially aneuploidy. An understanding of the regulation of meiosis, coupled with advances in genomics, may ultimately allow us to diagnose the causes of meiosis-based infertilities, more wisely apply assisted reproductive technologies, and derive functional germ cells.
Collapse
|
42
|
Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 1: Background to spermatogenesis, spermatogonia, and spermatocytes. Microsc Res Tech 2009; 73:241-78. [DOI: 10.1002/jemt.20783] [Citation(s) in RCA: 320] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
Wojtasz L, Daniel K, Roig I, Bolcun-Filas E, Xu H, Boonsanay V, Eckmann CR, Cooke HJ, Jasin M, Keeney S, McKay MJ, Toth A. Mouse HORMAD1 and HORMAD2, two conserved meiotic chromosomal proteins, are depleted from synapsed chromosome axes with the help of TRIP13 AAA-ATPase. PLoS Genet 2009; 5:e1000702. [PMID: 19851446 PMCID: PMC2758600 DOI: 10.1371/journal.pgen.1000702] [Citation(s) in RCA: 320] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Accepted: 09/25/2009] [Indexed: 11/18/2022] Open
Abstract
Meiotic crossovers are produced when programmed double-strand breaks (DSBs) are repaired by recombination from homologous chromosomes (homologues). In a wide variety of organisms, meiotic HORMA-domain proteins are required to direct DSB repair towards homologues. This inter-homologue bias is required for efficient homology search, homologue alignment, and crossover formation. HORMA-domain proteins are also implicated in other processes related to crossover formation, including DSB formation, inhibition of promiscuous formation of the synaptonemal complex (SC), and the meiotic prophase checkpoint that monitors both DSB processing and SCs. We examined the behavior of two previously uncharacterized meiosis-specific mouse HORMA-domain proteins—HORMAD1 and HORMAD2—in wild-type mice and in mutants defective in DSB processing or SC formation. HORMADs are preferentially associated with unsynapsed chromosome axes throughout meiotic prophase. We observe a strong negative correlation between SC formation and presence of HORMADs on axes, and a positive correlation between the presumptive sites of high checkpoint-kinase ATR activity and hyper-accumulation of HORMADs on axes. HORMADs are not depleted from chromosomes in mutants that lack SCs. In contrast, DSB formation and DSB repair are not absolutely required for depletion of HORMADs from synapsed axes. A simple interpretation of these findings is that SC formation directly or indirectly promotes depletion of HORMADs from chromosome axes. We also find that TRIP13 protein is required for reciprocal distribution of HORMADs and the SYCP1/SC-component along chromosome axes. Similarities in mouse and budding yeast meiosis suggest that TRIP13/Pch2 proteins have a conserved role in establishing mutually exclusive HORMAD-rich and synapsed chromatin domains in both mouse and yeast. Taken together, our observations raise the possibility that involvement of meiotic HORMA-domain proteins in the regulation of homologue interactions is conserved in mammals. Generation of haploid gametes in most organisms requires that homologues become connected via crossovers during meiosis. Efficient formation of crossovers depends on HORMA-domain proteins in diverse taxa. These proteins ensure that programmed meiotic DSBs are preferentially repaired from homologues, rather than from sister chromatids. This inter-homologue bias is crucial for homology search and crossovers formation. HORMA-domain proteins have been also implicated in DSB formation, in suppression of synaptonemal complex formation between non-homologous chromosomes, and in the meiotic prophase checkpoint that monitors DSB repair. Despite the importance of HORMA-domain proteins in various organisms, a role for these proteins in mammalian meiosis hasn't been reported. We examined the behaviour of meiotic mouse HORMA-domain proteins—HORMAD1 and HORMAD2—in wild-type and meiotic mutants. HORMAD1/2 preferentially accumulate on unsynapsed chromosome axes. Our data suggest that HORMAD1/2 depletion from chromosomes is a response to synaptonemal complex formation and it that is a conserved process supported by TRIP13/Pch2 AAA-ATPase. Assuming that HORMA-domain functions are conserved in mammals, we speculate that depletion of HORMADs from axes might contribute to the down-regulation of inter-homologue bias and the prophase checkpoint once homology search is completed and synaptonemal complexes form between aligned homologues.
Collapse
Affiliation(s)
- Lukasz Wojtasz
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Katrin Daniel
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Ignasi Roig
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | - Huiling Xu
- Divisions of Radiation Oncology and Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Verawan Boonsanay
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | | | - Howard J. Cooke
- Medical Research Council Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Howard Hughes Medical Institute, New York, New York, United States of America
| | - Michael J. McKay
- Department of Radiation Oncology, Australian National University and the Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Attila Toth
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
44
|
Adelfalk C, Janschek J, Revenkova E, Blei C, Liebe B, Göb E, Alsheimer M, Benavente R, de Boer E, Novak I, Höög C, Scherthan H, Jessberger R. Cohesin SMC1beta protects telomeres in meiocytes. J Cell Biol 2009; 187:185-99. [PMID: 19841137 PMCID: PMC2768837 DOI: 10.1083/jcb.200808016] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 09/17/2009] [Indexed: 12/29/2022] Open
Abstract
Meiosis-specific mammalian cohesin SMC1beta is required for complete sister chromatid cohesion and proper axes/loop structure of axial elements (AEs) and synaptonemal complexes (SCs). During prophase I, telomeres attach to the nuclear envelope (NE), but in Smc1beta(-/-) meiocytes, one fifth of their telomeres fail to attach. This study reveals that SMC1beta serves a specific role at telomeres, which is independent of its role in determining AE/SC length and loop extension. SMC1beta is necessary to prevent telomere shortening, and SMC3, present in all known cohesin complexes, properly localizes to telomeres only if SMC1beta is present. Very prominently, telomeres in Smc1beta(-/-) spermatocytes and oocytes loose their structural integrity and suffer a range of abnormalities. These include disconnection from SCs and formation of large telomeric protein-DNA extensions, extended telomere bridges between SCs, ring-like chromosomes, intrachromosomal telomeric repeats, and a reduction of SUN1 foci in the NE. We suggest that a telomere structure protected from DNA rearrangements depends on SMC1beta.
Collapse
Affiliation(s)
- Caroline Adelfalk
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Johannes Janschek
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Ekaterina Revenkova
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Cornelia Blei
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Bodo Liebe
- Max Planck Institute of Molecular Genetics, D-14195 Berlin, Germany
| | - Eva Göb
- Department of Cell and Developmental Biology, University of Würzburg, 97074 Würzburg, Germany
| | - Manfred Alsheimer
- Department of Cell and Developmental Biology, University of Würzburg, 97074 Würzburg, Germany
| | - Ricardo Benavente
- Department of Cell and Developmental Biology, University of Würzburg, 97074 Würzburg, Germany
| | - Esther de Boer
- Memorial Sloan-Kettering Cancer Center, New York, NY 10044
| | - Ivana Novak
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Christer Höög
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Harry Scherthan
- Max Planck Institute of Molecular Genetics, D-14195 Berlin, Germany
| | - Rolf Jessberger
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
45
|
Schoenmakers S, Wassenaar E, Hoogerbrugge JW, Laven JSE, Grootegoed JA, Baarends WM. Female meiotic sex chromosome inactivation in chicken. PLoS Genet 2009; 5:e1000466. [PMID: 19461881 PMCID: PMC2678266 DOI: 10.1371/journal.pgen.1000466] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Accepted: 04/03/2009] [Indexed: 12/25/2022] Open
Abstract
During meiotic prophase in male mammals, the heterologous X and Y chromosomes remain largely unsynapsed, and meiotic sex chromosome inactivation (MSCI) leads to formation of the transcriptionally silenced XY body. In birds, the heterogametic sex is female, carrying Z and W chromosomes (ZW), whereas males have the homogametic ZZ constitution. During chicken oogenesis, the heterologous ZW pair reaches a state of complete heterologous synapsis, and this might enable maintenance of transcription of Z- and W chromosomal genes during meiotic prophase. Herein, we show that the ZW pair is transiently silenced, from early pachytene to early diplotene using immunocytochemistry and gene expression analyses. We propose that ZW inactivation is most likely achieved via spreading of heterochromatin from the W on the Z chromosome. Also, persistent meiotic DNA double-strand breaks (DSBs) may contribute to silencing of Z. Surprisingly, gammaH2AX, a marker of DSBs, and also the earliest histone modification that is associated with XY body formation in mammalian and marsupial spermatocytes, does not cover the ZW during the synapsed stage. However, when the ZW pair starts to desynapse, a second wave of gammaH2AX accumulates on the unsynapsed regions of Z, which also show a reappearance of the DSB repair protein RAD51. This indicates that repair of meiotic DSBs on the heterologous part of Z is postponed until late pachytene/diplotene, possibly to avoid recombination with regions on the heterologously synapsed W chromosome. Two days after entering diplotene, the Z looses gammaH2AX and shows reactivation. This is the first report of meiotic sex chromosome inactivation in a species with female heterogamety, providing evidence that this mechanism is not specific to spermatogenesis. It also indicates the presence of an evolutionary force that drives meiotic sex chromosome inactivation independent of the final achievement of synapsis.
Collapse
Affiliation(s)
- Sam Schoenmakers
- Department of Reproduction and Development, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Evelyne Wassenaar
- Department of Reproduction and Development, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jos W. Hoogerbrugge
- Department of Reproduction and Development, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Joop S. E. Laven
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - J. Anton Grootegoed
- Department of Reproduction and Development, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Willy M. Baarends
- Department of Reproduction and Development, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
46
|
Recent advance in our understanding of the molecular nature of chromosomal abnormalities. J Hum Genet 2009; 54:253-60. [PMID: 19373258 DOI: 10.1038/jhg.2009.35] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The completion of the human genome project has enabled researchers to characterize the breakpoints for various chromosomal structural abnormalities including deletions, duplications or translocations. This in turn has shed new light on the molecular mechanisms underlying the onset of gross chromosomal rearrangements. On the other hand, advances in genetic manipulation technologies for various model organisms has increased our knowledge of meiotic chromosome segregation, errors which, contribute to chromosomal aneuploidy. This review focuses on the current understanding of germ line chromosomal abnormalities and provides an overview of the mechanisms involved. We refer to our own recent data and those of others to illustrate some of the new paradigms that have arisen in this field. We also discuss some perspectives on the sexual dimorphism of some of the pathways that leads to these chromosomal abnormalities.
Collapse
|
47
|
Bolor H, Mori T, Nishiyama S, Ito Y, Hosoba E, Inagaki H, Kogo H, Ohye T, Tsutsumi M, Kato T, Tong M, Nishizawa H, Pryor-Koishi K, Kitaoka E, Sawada T, Nishiyama Y, Udagawa Y, Kurahashi H. Mutations of the SYCP3 gene in women with recurrent pregnancy loss. Am J Hum Genet 2009; 84:14-20. [PMID: 19110213 DOI: 10.1016/j.ajhg.2008.12.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 11/19/2008] [Accepted: 12/04/2008] [Indexed: 12/17/2022] Open
Abstract
Aneuploidy, a chromosomal numerical abnormality in the conceptus or fetus, occurs in at least 5% of all pregnancies and is the leading cause of early pregnancy loss in humans. Accumulating evidence now suggests that the correct segregation of chromosomes is affected by events occurring in prophase during meiosis I. These events include homologous chromosome pairing, sister-chromatid cohesion, and meiotic recombination. In our current study, we show that mutations in SYCP3, a gene encoding an essential component of the synaptonemal complex that is central to the interaction of homologous chromosomes, are associated with recurrent pregnancy loss. Two out of 26 women with recurrent pregnancy loss of unknown cause were found to carry independent heterozygous nucleotide alterations in this gene, neither of which was present among a group of 150 fertile women. Analysis of transcripts from minigenes harboring each of these two mutations revealed that both affected normal splicing, possibly resulting in the production of C-terminally mutated proteins. The mutant proteins were found to interact with their wild-type counterpart in vitro and inhibit the normal fiber formation of the SYCP3 protein when coexpressed in a heterologous system. These data suggest that these mutations are likely to generate an aberrant synaptonemal complex in a dominant-negative manner and contribute to abnormal chromosomal behavior that might lead to recurrent miscarriage. Combined with the fact that similar mutations have been previously identified in two males with azoospermia, our current data suggest that sexual dimorphism in response to meiotic disruption occurs even in humans.
Collapse
|
48
|
Abstract
Accurate segregation of chromosomes during meiosis requires physical links between homologs. These links are usually established through chromosome pairing, synapsis, and recombination, which occur during meiotic prophase. How chromosomes pair with their homologous partners is one of the outstanding mysteries of meiosis. Surprisingly, experimental evidence indicates that different organisms have found more than one way to accomplish this feat. Whereas some species depend on recombination machinery to achieve homologous pairing, others are able to pair and synapse their homologs in the absence of recombination. To ensure specific pairing between homologous chromosomes, both recombination-dependent and recombination-independent mechanisms must strike the proper balance between forces that promote chromosome interactions and activities that temper the promiscuity of those interactions. The initiation of synapsis is likely to be a tightly regulated step in a process that must be mechanically coupled to homolog pairing.
Collapse
Affiliation(s)
- Needhi Bhalla
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California 95064, USA.
| | | |
Collapse
|
49
|
Protein SYCP2 provides a link between transverse filaments and lateral elements of mammalian synaptonemal complexes. Chromosoma 2008; 118:259-67. [PMID: 19034475 DOI: 10.1007/s00412-008-0194-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 10/30/2008] [Accepted: 11/03/2008] [Indexed: 01/22/2023]
Abstract
Synaptonemal complexes (SCs) are evolutionarily conserved meiosis-specific nuclear structures critically involved in synapsis, recombination, and segregation of homologous chromosomes. SCs are proteinaceous structures composed of (a) two lateral elements (LEs), to which the chromatin of the homologs is attached, (b) numerous transverse filaments (TFs) that link the LEs, and (c) a central element (CE). Major protein components of mammalian SCs are the TF protein SYCP1 and the LE proteins SYCP2 and SCYP3. How SCs become assembled is presently poorly understood, in particular, it is not known how TFs assemble at the plane of LEs to interconnect the homologous chromosomes. Therefore, we have investigated possible interactions between SYCP1 and other SC proteins. In immunoprecipitation experiments we could find that SYCP1 and SYCP2 interact in extracts of meiotic cells. Using the yeast two-hybrid system, we were able to demonstrate that the C-terminus of SYCP1 directly interacts with SYCP2. These results were confirmed by different interaction traps. Furthermore, we could narrow down the interacting domain of the SYCP2 molecule to its C-terminal region. We propose that SYCP2 acts as a linker between SYCP1 and SYCP3 and therefore would be the missing connecting link between LEs and TFs essential for proper chromosome synapsis.
Collapse
|
50
|
Öllinger R, Childs AJ, Burgess HM, Speed RM, Lundegaard PR, Reynolds N, Gray NK, Cooke HJ, Adams IR. Deletion of the pluripotency-associated Tex19.1 gene causes activation of endogenous retroviruses and defective spermatogenesis in mice. PLoS Genet 2008; 4:e1000199. [PMID: 18802469 PMCID: PMC2531233 DOI: 10.1371/journal.pgen.1000199] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Accepted: 08/15/2008] [Indexed: 01/06/2023] Open
Abstract
As genetic information is transmitted through successive generations, it passes between pluripotent cells in the early embryo and germ cells in the developing foetus and adult animal. Tex19.1 encodes a protein of unknown function, whose expression is restricted to germ cells and pluripotent cells. During male spermatogenesis, Tex19.1 expression is highest in mitotic spermatogonia and diminishes as these cells differentiate and progress through meiosis. In pluripotent stem cells, Tex19.1 expression is also downregulated upon differentiation. However, it is not clear whether Tex19.1 has an essential function in germ cells or pluripotent stem cells, or what that function might be. To analyse the potential role of Tex19.1 in pluripotency or germ cell function we have generated Tex19.1(-/-) knockout mice and analysed the Tex19.1(-/-) mutant phenotype. Adult Tex19.1(-/-) knockout males exhibit impaired spermatogenesis. Immunostaining and histological analysis revealed defects in meiotic chromosome synapsis, the persistence of DNA double-strand breaks during meiosis, and a loss of post-meiotic germ cells in the testis. Furthermore, expression of a class of endogenous retroviruses is upregulated during meiosis in the Tex19.1(-/-) testes. Increased transposition of endogenous retroviruses in the germline of Tex19.1(-/-) mutant mice, and the concomitant increase in DNA damage, may be sufficient to disrupt the normal processes of recombination and chromosome synapsis during meiosis and cause defects in spermatogenesis. Our results suggest that Tex19.1 is part of a specialised mechanism that operates in the germline to repress transposable genetic elements and maintain genomic stability through successive generations.
Collapse
Affiliation(s)
- Rupert Öllinger
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Andrew J. Childs
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Hannah M. Burgess
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- School of Clinical Sciences and Community Health, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Robert M. Speed
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Pia R. Lundegaard
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- Institute for Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Nicola Reynolds
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Nicola K. Gray
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- School of Clinical Sciences and Community Health, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Howard J. Cooke
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
| | - Ian R. Adams
- MRC Human Genetics Unit, Western General Hospital, Edinburgh, United Kingdom
- Edinburgh Cancer Research Centre, School of Molecular and Clinical Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| |
Collapse
|