Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 84(10): 1826-1841.e5, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38657614

RESUMO

In meiotic cells, chromosomes are organized as chromatin loop arrays anchored to a protein axis. This organization is essential to regulate meiotic recombination, from DNA double-strand break (DSB) formation to their repair. In mammals, it is unknown how chromatin loops are organized along the genome and how proteins participating in DSB formation are tethered to the chromosome axes. Here, we identify three categories of axis-associated genomic sites: PRDM9 binding sites, where DSBs form; binding sites of the insulator protein CTCF; and H3K4me3-enriched sites. We demonstrate that PRDM9 promotes the recruitment of MEI4 and IHO1, two proteins essential for DSB formation. In turn, IHO1 anchors DSB sites to the axis components HORMAD1 and SYCP3. We discovered that IHO1, HORMAD1, and SYCP3 are associated at the DSB ends during DSB repair. Our results highlight how interactions of proteins with specific genomic elements shape the meiotic chromosome organization for recombination.


Assuntos
Quebras de DNA de Cadeia Dupla , Histona-Lisina N-Metiltransferase , Meiose , Meiose/genética , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Camundongos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Histonas/metabolismo , Histonas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Sítios de Ligação , Cromossomos/genética , Cromossomos/metabolismo , Cromatina/metabolismo , Cromatina/genética , Fator de Ligação a CCCTC/metabolismo , Fator de Ligação a CCCTC/genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Recombinação Genética , Masculino
2.
Genes Dev ; 36(1-2): 4-6, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-35022326

RESUMO

During meiosis, a molecular program induces DNA double-strand breaks (DSBs) and their repair by homologous recombination. DSBs can be repaired with or without crossovers. ZMM proteins promote the repair toward crossover. The sites of DSB repair are also sites where the axes of homologous chromosomes are juxtaposed and stabilized, and where a structure called the synaptonemal complex initiates, providing further regulation of both DSB formation and repair. How crossover formation and synapsis initiation are linked has remained unknown. The study by Pyatnitskaya and colleagues (pp. 53-69) in this issue of Genes & Development highlights the central role of the Saccharomyces cerevisiae ZMM protein Zip4 in this process.


Assuntos
Troca Genética , Complexo Sinaptonêmico , Pareamento Cromossômico , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Meiose/genética
3.
Mol Cell ; 74(5): 1069-1085.e11, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31000436

RESUMO

Orderly segregation of chromosomes during meiosis requires that crossovers form between homologous chromosomes by recombination. Programmed DNA double-strand breaks (DSBs) initiate meiotic recombination. We identify ANKRD31 as a key component of complexes of DSB-promoting proteins that assemble on meiotic chromosome axes. Genome-wide, ANKRD31 deficiency causes delayed recombination initiation. In addition, loss of ANKRD31 alters DSB distribution because of reduced selectivity for sites that normally attract DSBs. Strikingly, ANKRD31 deficiency also abolishes uniquely high rates of recombination that normally characterize pseudoautosomal regions (PARs) of X and Y chromosomes. Consequently, sex chromosomes do not form crossovers, leading to chromosome segregation failure in ANKRD31-deficient spermatocytes. These defects co-occur with a genome-wide delay in assembling DSB-promoting proteins on autosome axes and loss of a specialized PAR-axis domain that is highly enriched for DSB-promoting proteins in wild type. Thus, we propose a model for spatiotemporal patterning of recombination by ANKRD31-dependent control of axis-associated DSB-promoting proteins.


Assuntos
Proteínas de Transporte/genética , Quebras de DNA de Cadeia Dupla , Recombinação Homóloga/genética , Meiose/genética , Animais , Proteínas de Transporte/química , Segregação de Cromossomos/genética , Masculino , Camundongos , Regiões Pseudoautossômicas/genética , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Cromossomo X/genética , Cromossomo Y/genética
4.
BMC Biol ; 19(1): 86, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33910563

RESUMO

BACKGROUND: Vertebrate meiotic recombination events are concentrated in regions (hotspots) that display open chromatin marks, such as trimethylation of lysines 4 and 36 of histone 3 (H3K4me3 and H3K36me3). Mouse and human PRDM9 proteins catalyze H3K4me3 and H3K36me3 and determine hotspot positions, whereas other vertebrates lacking PRDM9 recombine in regions with chromatin already opened for another function, such as gene promoters. While these other vertebrate species lacking PRDM9 remain fertile, inactivation of the mouse Prdm9 gene, which shifts the hotspots to the functional regions (including promoters), typically causes gross fertility reduction; and the reasons for these species differences are not clear. RESULTS: We introduced Prdm9 deletions into the Rattus norvegicus genome and generated the first rat genome-wide maps of recombination-initiating double-strand break hotspots. Rat strains carrying the same wild-type Prdm9 allele shared 88% hotspots but strains with different Prdm9 alleles only 3%. After Prdm9 deletion, rat hotspots relocated to functional regions, about 40% to positions corresponding to Prdm9-independent mouse hotspots, including promoters. Despite the hotspot relocation and decreased fertility, Prdm9-deficient rats of the SHR/OlaIpcv strain produced healthy offspring. The percentage of normal pachytene spermatocytes in SHR-Prdm9 mutants was almost double than in the PWD male mouse oligospermic sterile mutants. We previously found a correlation between the crossover rate and sperm presence in mouse Prdm9 mutants. The crossover rate of SHR is more similar to sperm-carrying mutant mice, but it did not fully explain the fertility of the SHR mutants. Besides mild meiotic arrests at rat tubular stages IV (mid-pachytene) and XIV (metaphase), we also detected postmeiotic apoptosis of round spermatids. We found delayed meiosis and age-dependent fertility in both sexes of the SHR mutants. CONCLUSIONS: We hypothesize that the relative increased fertility of rat versus mouse Prdm9 mutants could be ascribed to extended duration of meiotic prophase I. While rat PRDM9 shapes meiotic recombination landscapes, it is unnecessary for recombination. We suggest that PRDM9 has additional roles in spermatogenesis and speciation-spermatid development and reproductive age-that may help to explain male-specific hybrid sterility.


Assuntos
Meiose , Animais , Cromatina , Quebras de DNA de Cadeia Dupla , Feminino , Fertilidade/genética , Histona-Lisina N-Metiltransferase/genética , Masculino , Meiose/genética , Camundongos , Ratos , Ratos Endogâmicos SHR , Espermatogênese/genética
5.
PLoS Genet ; 14(8): e1007479, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30161134

RESUMO

During meiosis, maternal and paternal chromosomes undergo exchanges by homologous recombination. This is essential for fertility and contributes to genome evolution. In many eukaryotes, sites of meiotic recombination, also called hotspots, are regions of accessible chromatin, but in many vertebrates, their location follows a distinct pattern and is specified by PR domain-containing protein 9 (PRDM9). The specification of meiotic recombination hotspots is achieved by the different activities of PRDM9: DNA binding, histone methyltransferase, and interaction with other proteins. Remarkably, PRDM9 activity leads to the erosion of its own binding sites and the rapid evolution of its DNA-binding domain. PRDM9 may also contribute to reproductive isolation, as it is involved in hybrid sterility potentially due to a reduction of its activity in specific heterozygous contexts.


Assuntos
Mapeamento Cromossômico , Histona-Lisina N-Metiltransferase/genética , Meiose , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ligação a DNA , Evolução Molecular , Fertilidade , Heterozigoto , Histona-Lisina N-Metiltransferase/metabolismo , Recombinação Homóloga , Humanos , Infertilidade , Masculino , Camundongos , Conformação Proteica , Isolamento Reprodutivo , Espermatócitos
6.
Genome Res ; 27(4): 580-590, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28336543

RESUMO

In mouse and human meiosis, DNA double-strand breaks (DSBs) initiate homologous recombination and occur at specific sites called hotspots. The localization of these sites is determined by the sequence-specific DNA binding domain of the PRDM9 histone methyl transferase. Here, we performed an extensive analysis of PRDM9 binding in mouse spermatocytes. Unexpectedly, we identified a noncanonical recruitment of PRDM9 to sites that lack recombination activity and the PRDM9 binding consensus motif. These sites include gene promoters, where PRDM9 is recruited in a DSB-dependent manner. Another subset reveals DSB-independent interactions between PRDM9 and genomic sites, such as the binding sites for the insulator protein CTCF. We propose that these DSB-independent sites result from interactions between hotspot-bound PRDM9 and genomic sequences located on the chromosome axis.


Assuntos
Genoma , Histona-Lisina N-Metiltransferase/metabolismo , Motivos de Nucleotídeos , Animais , Fator de Ligação a CCCTC/metabolismo , Quebras de DNA de Cadeia Dupla , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Ligação Proteica , Espermatócitos/metabolismo
7.
Mol Cell ; 47(4): 523-34, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22771120

RESUMO

We generated knockout mice for MCM8 and MCM9 and show that deficiency for these genes impairs homologous recombination (HR)-mediated DNA repair during gametogenesis and somatic cells cycles. MCM8(-/-) mice are sterile because spermatocytes are blocked in meiotic prophase I, and females have only arrested primary follicles and frequently develop ovarian tumors. MCM9(-/-) females also are sterile as ovaries are completely devoid of oocytes. In contrast, MCM9(-/-) testes produce spermatozoa, albeit in much reduced quantity. Mcm8(-/-) and Mcm9(-/-) embryonic fibroblasts show growth defects and chromosomal damage and cannot overcome a transient inhibition of replication fork progression. In these cells, chromatin recruitment of HR factors like Rad51 and RPA is impaired and HR strongly reduced. We further demonstrate that MCM8 and MCM9 form a complex and that they coregulate their stability. Our work uncovers essential functions of MCM8 and MCM9 in HR-mediated DSB repair during gametogenesis, replication fork maintenance, and DNA repair.


Assuntos
Proteínas de Ciclo Celular/deficiência , Proteínas de Ligação a DNA/deficiência , Gametogênese/genética , Instabilidade Genômica , Recombinação Homóloga/genética , Animais , Proteínas de Ciclo Celular/genética , Cromatina/genética , Reparo do DNA , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/metabolismo , Células Germinativas/metabolismo , Masculino , Prófase Meiótica I/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Manutenção de Minicromossomo , Ovário/metabolismo , Espermatócitos/metabolismo
8.
Reproduction ; 151(3): 239-51, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26667018

RESUMO

In sexually reproducing organisms, accurate gametogenesis is crucial for the transmission of genetic material from one generation to the next. This requires the faithful segregation of chromosomes during mitotic and meiotic divisions. One of the main players in this process is the kinetochore, a large multi-protein complex that forms at the interface of centromeres and microtubules. Here, we analyzed the expression profile and function of small kinetochore-associated protein (SKAP) in the mouse. We found that two distinct SKAP isoforms are specifically expressed in the germline: a smaller isoform, which is detected in spermatogonia and spermatocytes and localized in the outer mitotic and meiotic kinetochores from metaphase to telophase, and a larger isoform, which is expressed in the cytoplasm of elongating spermatids. We generated SKAP-deficient mice and found that testis size and sperm production were severely reduced in mutant males. This phenotype was partially caused by defects during spermatogonia proliferation before entry into meiosis. We conclude that mouse SKAP, while being dispensable for somatic cell divisions, has an important role in the successful outcome of male gametogenesis. In germ cells, analogous to what has been suggested in studies using immortalized cells, SKAP most likely stabilizes the interaction between kinetochores and microtubules, where it might be needed as an extra safeguard to ensure the correct segregation of mitotic and meiotic chromosomes.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Animais , Apoptose , Feminino , Fertilidade , Células HeLa , Humanos , Masculino , Meiose , Camundongos , Mitose , Espermatozoides/crescimento & desenvolvimento
9.
PLoS Biol ; 9(10): e1001176, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028627

RESUMO

Meiotic recombination generates reciprocal exchanges between homologous chromosomes (also called crossovers, COs) that are essential for proper chromosome segregation during meiosis and are a major source of genome diversity by generating new allele combinations. COs have two striking properties: they occur at specific sites, called hotspots, and these sites evolve rapidly. In mammals, the Prdm9 gene, which encodes a meiosis-specific histone H3 methyltransferase, has recently been identified as a determinant of CO hotspots. Here, using transgenic mice, we show that the sole modification of PRDM9 zinc fingers leads to changes in hotspot activity, histone H3 lysine 4 trimethylation (H3K4me3) levels, and chromosome-wide distribution of COs. We further demonstrate by an in vitro assay that the PRDM9 variant associated with hotspot activity binds specifically to DNA sequences located at the center of the three hotspots tested. Remarkably, we show that mutations in cis located at hotspot centers and associated with a decrease of hotspot activity affect PRDM9 binding. Taken together, these results provide the direct demonstration that Prdm9 is a master regulator of hotspot localization through the DNA binding specificity of its zinc finger array and that binding of PRDM9 at hotspots promotes local H3K4me3 enrichment.


Assuntos
Troca Genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Meiose , Dedos de Zinco , Animais , Quebras de DNA de Cadeia Dupla , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
10.
EMBO J ; 28(17): 2616-24, 2009 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-19644444

RESUMO

Little is known about the factors determining the location and activity of the rapidly evolving meiotic crossover hotspots that shape genome diversity. Here, we show that several histone modifications are enriched at the active mouse Psmb9 hotspot, and we distinguish those marks that precede from those that follow hotspot recombinational activity. H3K4Me3, H3K4Me2 and H3K9Ac are specifically enriched in the chromatids that carry an active initiation site, and in the absence of DNA double-strand breaks (DSBs) in Spo11(-/-) mice. We thus propose that these marks are part of the substrate for recombination initiation at the Psmb9 hotspot. In contrast, hyperacetylation of H4 is increased as a consequence of DSB formation, as shown by its dependency on Spo11 and by the enrichment detected on both recombining chromatids. In addition, the comparison with another hotspot, Hlx1, strongly suggests that H3K4Me3 and H4 hyperacetylation are common features of DSB formation and repair, respectively. Altogether, the chromatin signatures of the Psmb9 and Hlx1 hotspots provide a basis for understanding the distribution of meiotic recombination.


Assuntos
Histonas/metabolismo , Meiose/genética , Recombinação Genética , Animais , Troca Genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Camundongos , Camundongos Endogâmicos
11.
PLoS Biol ; 7(2): e35, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19226188

RESUMO

Meiotic recombination events are not randomly distributed in the genome but occur in specific regions called recombination hotspots. Hotspots are predicted to be preferred sites for the initiation of meiotic recombination and their positions and activities are regulated by yet-unknown controls. The activity of the Psmb9 hotspot on mouse Chromosome 17 (Chr 17) varies according to genetic background. It is active in strains carrying a recombinant Chr 17 where the proximal third is derived from Mus musculus molossinus. We have identified the genetic locus required for Psmb9 activity, named Dsbc1 for Double-strand break control 1, and mapped this locus within a 6.7-Mb region on Chr 17. Based on cytological analysis of meiotic DNA double-strand breaks (DSB) and crossovers (COs), we show that Dsbc1 influences DSB and CO, not only at Psmb9, but in several other regions of Chr 17. We further show that CO distribution is also influenced by Dsbc1 on Chrs 15 and 18. Finally, we provide direct molecular evidence for the regulation in trans mediated by Dsbc1, by showing that it controls the CO activity at the Hlx1 hotspot on Chr 1. We thus propose that Dsbc1 encodes for a trans-acting factor involved in the specification of initiation sites of meiotic recombination genome wide in mice.


Assuntos
Estudo de Associação Genômica Ampla , Genoma , Meiose/genética , Camundongos Endogâmicos/genética , Recombinação Genética/genética , Animais , Núcleo Celular/química , Núcleo Celular/metabolismo , Mapeamento Cromossômico , Cromossomos de Mamíferos , Troca Genética , Cisteína Endopeptidases/genética , Quebras de DNA de Cadeia Dupla , Hibridização in Situ Fluorescente , Masculino , Camundongos , Repetições de Microssatélites , Espermatócitos/química , Espermatócitos/citologia , Espermatócitos/metabolismo
12.
PLoS Genet ; 5(2): e1000393, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19247432

RESUMO

In mammals, the synaptonemal complex is a structure required to complete crossover recombination. Although suggested by cytological work, in vivo links between the structural proteins of the synaptonemal complex and the proteins of the recombination process have not previously been made. The central element of the synaptonemal complex is traversed by DNA at sites of recombination and presents a logical place to look for interactions between these components. There are four known central element proteins, three of which have previously been mutated. Here, we complete the set by creating a null mutation in the Syce1 gene in mouse. The resulting disruption of synapsis in these animals has allowed us to demonstrate a biochemical interaction between the structural protein SYCE2 and the repair protein RAD51. In normal meiosis, this interaction may be responsible for promoting homologous synapsis from sites of recombination.


Assuntos
Pareamento Cromossômico , Reparo do DNA , Proteínas Nucleares/genética , Complexo Sinaptonêmico/metabolismo , Animais , Feminino , Gametogênese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Ligação Proteica , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Recombinação Genética , Espermatócitos/citologia , Espermatócitos/metabolismo , Complexo Sinaptonêmico/genética
13.
Nat Commun ; 13(1): 7048, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396648

RESUMO

Meiosis requires the formation of programmed DNA double strand breaks (DSBs), essential for fertility and for generating genetic diversity. DSBs are induced by the catalytic activity of the TOPOVIL complex formed by SPO11 and TOPOVIBL. To ensure genomic integrity, DNA cleavage activity is tightly regulated, and several accessory factors (REC114, MEI4, IHO1, and MEI1) are needed for DSB formation in mice. How and when these proteins act is not understood. Here, we show that REC114 is a direct partner of TOPOVIBL, and identify their conserved interacting domains by structural analysis. We then analyse the role of this interaction by monitoring meiotic DSBs in female and male mice carrying point mutations in TOPOVIBL that decrease or disrupt its binding to REC114. In these mutants, DSB activity is strongly reduced genome-wide in oocytes, and only in sub-telomeric regions in spermatocytes. In addition, in mutant spermatocytes, DSB activity is delayed in autosomes. These results suggest that REC114 is a key member of the TOPOVIL catalytic complex, and that the REC114/TOPOVIBL interaction ensures the efficiency and timing of DSB activity.


Assuntos
Quebras de DNA de Cadeia Dupla , Meiose , Masculino , Feminino , Camundongos , Animais , Meiose/genética , Cromossomos , Espermatócitos , DNA
14.
Dev Cell ; 11(4): 535-46, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17011492

RESUMO

The transcriptional mechanisms underlying lineage specification and differentiation of embryonic stem (ES) cells remain elusive. Oct-3/4 (POU5f1) is one of the earliest transcription factors expressed in the embryo. Both the pluripotency and the fate of ES cells depend upon a tight control of Oct-3/4 expression. We report that transgene- or TGFbeta-induced increase in Oct-3/4 mRNA and protein levels in undifferentiated ES cells and at early stages of differentiation triggers expression of mesodermal and cardiac specific genes through Smad2/4. cDNA antisense- and siRNA-mediated inhibition of upregulation of Oct-3/4 in ES cells prevent their specification toward the mesoderm and their differentiation into cardiomyocytes. Similarly, Oct-3/4 siRNA injected in the inner cell mass of blastocysts impairs cardiogenesis in early embryos. Thus, quantitative Oct-3/4 expression is regulated by a morphogen, pointing to a pivotal and physiological function of the POU factor in mesodermal and cardiac commitments of ES cells and of the epiblast.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/crescimento & desenvolvimento , Miócitos Cardíacos/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Linhagem da Célula , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Microinjeções , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Técnicas de Cultura de Órgãos , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/metabolismo
15.
Med Sci (Paris) ; 27(1): 63-9, 2011 Jan.
Artigo em Francês | MEDLINE | ID: mdl-21299964

RESUMO

During meiosis, homologous reciprocal recombination events or crossing-over determine the genetic map and are known not to be randomly distributed in the genome. Recent studies in yeasts and mammals reveal some key features of the molecular mechanism involved in this distribution. Through different molecular processes, specific histone post-translational modifications are induced at specific genomic sites, called hotspots, where initiation of meiotic recombination takes place. These sites are some transcription promoters in S. cerevisiae or binding sites for transcription factors in S. pombe, where chromatin modifiers are recruited. In mammals, the sites are DNA sequences recognized by the PRDM9 protein which has the ability both to bind DNA and to induce the trimethylation of the lysine 4 of histone H3. The properties of the chromatin at these sites, and potentially the binding of additional factors, allow the recruitment of proteins involved in the formation of DNA double strand breaks that initiate meiotic recombination.


Assuntos
Cromossomos/genética , Troca Genética/genética , Meiose , Acetilação , Animais , Montagem e Desmontagem da Cromatina/fisiologia , Cromossomos Fúngicos/genética , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/fisiologia , Endodesoxirribonucleases/antagonistas & inibidores , Endodesoxirribonucleases/fisiologia , Proteínas Fúngicas/metabolismo , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/fisiologia , Histonas/metabolismo , Humanos , Mamíferos/genética , Metilação , Camundongos , Modelos Genéticos , Processamento de Proteína Pós-Traducional , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/fisiologia , Schizosaccharomyces/genética , Especificidade da Espécie , Fatores de Transcrição/fisiologia , Dedos de Zinco/genética
16.
Front Cell Dev Biol ; 9: 688878, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34150782

RESUMO

One of the most fascinating aspects of meiosis is the extensive reorganization of the genome at the prophase of the first meiotic division (prophase I). The first steps of this reorganization are observed with the establishment of an axis structure, that connects sister chromatids, from which emanate arrays of chromatin loops. This axis structure, called the axial element, consists of various proteins, such as cohesins, HORMA-domain proteins, and axial element proteins. In many organisms, axial elements are required to set the stage for efficient sister chromatid cohesion and meiotic recombination, necessary for the recognition of the homologous chromosomes. Here, we review the different actors involved in axial element formation in Saccharomyces cerevisiae and in mouse. We describe the current knowledge of their localization pattern during prophase I, their functional interdependence, their role in sister chromatid cohesion, loop axis formation, homolog pairing before meiotic recombination, and recombination. We also address further challenges that need to be resolved, to fully understand the interplay between the chromosome structure and the different molecular steps that take place in early prophase I, which lead to the successful outcome of meiosis I.

17.
Elife ; 92020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33047671

RESUMO

Meiotic recombination starts with the formation of DNA double-strand breaks (DSBs) at specific genomic locations that correspond to PRDM9-binding sites. The molecular steps occurring from PRDM9 binding to DSB formation are unknown. Using proteomic approaches to find PRDM9 partners, we identified HELLS, a member of the SNF2-like family of chromatin remodelers. Upon functional analyses during mouse male meiosis, we demonstrated that HELLS is required for PRDM9 binding and DSB activity at PRDM9 sites. However, HELLS is not required for DSB activity at PRDM9-independent sites. HELLS is also essential for 5-hydroxymethylcytosine (5hmC) enrichment at PRDM9 sites. Analyses of 5hmC in mice deficient for SPO11, which catalyzes DSB formation, and in PRDM9 methyltransferase deficient mice reveal that 5hmC is triggered at DSB-prone sites upon PRDM9 binding and histone modification, but independent of DSB activity. These findings highlight the complex regulation of the chromatin and epigenetic environments at PRDM9-specified hotspots.


Assuntos
5-Metilcitosina/análogos & derivados , Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Histona-Lisina N-Metiltransferase/genética , 5-Metilcitosina/metabolismo , Animais , Sítios de Ligação , Endodesoxirribonucleases/metabolismo , Células HeLa , Histona-Lisina N-Metiltransferase/metabolismo , Recombinação Homóloga , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteômica , Espermatócitos/citologia , Testículo/metabolismo
20.
Nat Genet ; 46(10): 1072-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25151354

RESUMO

The ability to examine all chromatids from a single meiosis in yeast tetrads has been indispensable for defining the mechanisms of homologous recombination initiated by DNA double-strand breaks (DSBs). Using a broadly applicable strategy for the analysis of chromatids from a single meiosis at two recombination hotspots in mouse oocytes and spermatocytes, we demonstrate here the unidirectional transfer of information-gene conversion-in both crossovers and noncrossovers. Whereas gene conversion in crossovers is associated with reciprocal exchange, the unbroken chromatid is not altered in noncrossover gene conversion events, providing strong evidence that noncrossovers arise from a distinct pathway. Gene conversion frequently spares the binding site of the hotspot-specifying protein PRDM9, with the result that erosion of the hotspot is slowed. Thus, mouse tetrad analysis demonstrates how unique aspects of mammalian recombination mechanisms shape hotspot evolutionary dynamics.


Assuntos
Evolução Molecular , Meiose/genética , Oócitos/metabolismo , Recombinação Genética/genética , Espermatócitos/metabolismo , Animais , Southwestern Blotting , Cromossomos de Mamíferos/genética , Cromossomos de Mamíferos/metabolismo , Troca Genética , Feminino , Conversão Gênica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Modelos Genéticos , Oócitos/citologia , Espermatócitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA