Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 84(10): 1826-1841.e5, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38657614

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , N-Metiltransferasa de Histona-Lisina , Meiosis , Meiosis/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Histonas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Sitios de Unión , Cromosomas/genética , Cromosomas/metabolismo , Cromatina/metabolismo , Cromatina/genética , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Recombinación Genética , Masculino
2.
Nat Commun ; 13(1): 7048, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396648

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , Meiosis , Masculino , Femenino , Ratones , Animales , Meiosis/genética , Cromosomas , Espermatocitos , ADN
3.
Genes Dev ; 36(1-2): 4-6, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35022326

RESUMEN

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.


Asunto(s)
Intercambio Genético , Complejo Sinaptonémico , Emparejamiento Cromosómico , Roturas del ADN de Doble Cadena , Reparación del ADN , Meiosis/genética
4.
Front Cell Dev Biol ; 9: 688878, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150782

RESUMEN

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.

5.
BMC Biol ; 19(1): 86, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33910563

RESUMEN

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.


Asunto(s)
Meiosis , Animales , Cromatina , Roturas del ADN de Doble Cadena , Femenino , Fertilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Masculino , Meiosis/genética , Ratones , Ratas , Ratas Endogámicas SHR , Espermatogénesis/genética
6.
Elife ; 92020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33047671

RESUMEN

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.


Asunto(s)
5-Metilcitosina/análogos & derivados , Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , 5-Metilcitosina/metabolismo , Animales , Sitios de Unión , Endodesoxirribonucleasas/metabolismo , Células HeLa , N-Metiltransferasa de Histona-Lisina/metabolismo , Recombinación Homóloga , Humanos , Masculino , Ratones , Ratones Noqueados , Proteómica , Espermatocitos/citología , Testículo/metabolismo
7.
Mol Cell ; 74(5): 1069-1085.e11, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000436

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Roturas del ADN de Doble Cadena , Recombinación Homóloga/genética , Meiosis/genética , Animales , Proteínas Portadoras/química , Segregación Cromosómica/genética , Masculino , Ratones , Regiones Pseudoautosómicas/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Cromosoma X/genética , Cromosoma Y/genética
8.
PLoS Genet ; 14(8): e1007479, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30161134

RESUMEN

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.


Asunto(s)
Mapeo Cromosómico , N-Metiltransferasa de Histona-Lisina/genética , Meiosis , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Unión al ADN , Evolución Molecular , Fertilidad , Heterocigoto , N-Metiltransferasa de Histona-Lisina/metabolismo , Recombinación Homóloga , Humanos , Infertilidad , Masculino , Ratones , Conformación Proteica , Aislamiento Reproductivo , Espermatocitos
9.
Genome Res ; 27(4): 580-590, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28336543

RESUMEN

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.


Asunto(s)
Genoma , N-Metiltransferasa de Histona-Lisina/metabolismo , Motivos de Nucleótidos , Animales , Factor de Unión a CCCTC/metabolismo , Roturas del ADN de Doble Cadena , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Espermatocitos/metabolismo
10.
Reproduction ; 151(3): 239-51, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26667018

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Animales , Apoptosis , Femenino , Fertilidad , Células HeLa , Humanos , Masculino , Meiosis , Ratones , Mitosis , Espermatozoides/crecimiento & desarrollo
11.
Nat Genet ; 46(10): 1072-80, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25151354

RESUMEN

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.


Asunto(s)
Evolución Molecular , Meiosis/genética , Oocitos/metabolismo , Recombinación Genética/genética , Espermatocitos/metabolismo , Animales , Southwestern Blotting , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Intercambio Genético , Femenino , Conversión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos , Modelos Genéticos , Oocitos/citología , Espermatocitos/citología
12.
Mol Cell ; 47(4): 523-34, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22771120

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Proteínas de Unión al ADN/deficiencia , Gametogénesis/genética , Inestabilidad Genómica , Recombinación Homóloga/genética , Animales , Proteínas de Ciclo Celular/genética , Cromatina/genética , Reparación del ADN , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/metabolismo , Células Germinativas/metabolismo , Masculino , Profase Meiótica I/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Mantenimiento de Minicromosoma , Ovario/metabolismo , Espermatocitos/metabolismo
14.
PLoS Biol ; 9(10): e1001176, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028627

RESUMEN

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.


Asunto(s)
Intercambio Genético , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Meiosis , Dedos de Zinc , Animales , Roturas del ADN de Doble Cadena , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
PLoS One ; 6(10): e25241, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21998645

RESUMEN

BACKGROUND: Spermatogenesis is a complex biological process that requires a highly specialized control of gene expression. In the past decade, small non-coding RNAs have emerged as critical regulators of gene expression both at the transcriptional and post-transcriptional level. DICER1, an RNAse III endonuclease, is essential for the biogenesis of several classes of small RNAs, including microRNAs (miRNAs) and endogenous small interfering RNAs (endo-siRNAs), but is also critical for the degradation of toxic transposable elements. In this study, we investigated to which extent DICER1 is required for germ cell development and the progress of spermatogenesis in mice. PRINCIPAL FINDINGS: We show that the selective ablation of Dicer1 at the early onset of male germ cell development leads to infertility, due to multiple cumulative defects at the meiotic and post-meiotic stages culminating with the absence of functional spermatozoa. Alterations were observed in the first spermatogenic wave and include delayed progression of spermatocytes to prophase I and increased apoptosis, resulting in a reduced number of round spermatids. The transition from round to mature spermatozoa was also severely affected, since the few spermatozoa formed in mutant animals were immobile and misshapen, exhibiting morphological defects of the head and flagellum. We also found evidence that the expression of transposable elements of the SINE family is up-regulated in Dicer1-depleted spermatocytes. CONCLUSIONS/SIGNIFICANCE: Our findings indicate that DICER1 is dispensable for spermatogonial stem cell renewal and mitotic proliferation, but is required for germ cell differentiation through the meiotic and haploid phases of spermatogenesis.


Asunto(s)
ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Infertilidad Masculina/genética , Meiosis/genética , Ribonucleasa III/deficiencia , Ribonucleasa III/genética , Espermatogénesis/genética , Espermatozoides/metabolismo , Espermatozoides/patología , Animales , Apoptosis/genética , Elementos Transponibles de ADN/genética , Eliminación de Gen , Silenciador del Gen , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Transgénicos , MicroARNs/genética , Tamaño de los Órganos/genética , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/patología , Recuento de Espermatozoides , Espermatocitos/metabolismo , Espermatocitos/patología
16.
Med Sci (Paris) ; 27(1): 63-9, 2011 Jan.
Artículo en Francés | MEDLINE | ID: mdl-21299964

RESUMEN

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.


Asunto(s)
Cromosomas/genética , Intercambio Genético/genética , Meiosis , Acetilación , Animales , Ensamble y Desensamble de Cromatina/fisiología , Cromosomas Fúngicos/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/fisiología , Endodesoxirribonucleasas/antagonistas & inhibidores , Endodesoxirribonucleasas/fisiología , Proteínas Fúngicas/metabolismo , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/metabolismo , Humanos , Mamíferos/genética , Metilación , Ratones , Modelos Genéticos , Procesamiento Proteico-Postraduccional , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/fisiología , Schizosaccharomyces/genética , Especificidad de la Especie , Factores de Transcripción/fisiología , Dedos de Zinc/genética
18.
PLoS One ; 5(12): e15770, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21209834

RESUMEN

The molecular chaperone Hsp90 has been found to be essential for viability in all tested eukaryotes, from the budding yeast to Drosophila. In mammals, two genes encode the two highly similar and functionally largely redundant isoforms Hsp90α and Hsp90ß. Although they are co-expressed in most if not all cells, their relative levels vary between tissues and during development. Since mouse embryos lacking Hsp90ß die at implantation, and despite the fact that Hsp90 inhibitors being tested as anti-cancer agents are relatively well tolerated, the organismic functions of Hsp90 in mammals remain largely unknown. We have generated mouse lines carrying gene trap insertions in the Hsp90α gene to investigate the global functions of this isoform. Surprisingly, mice without Hsp90α are apparently normal, with one major exception. Mutant male mice, whose Hsp90ß levels are unchanged, are sterile because of a complete failure to produce sperm. While the development of the male reproductive system appears to be normal, spermatogenesis arrests specifically at the pachytene stage of meiosis I. Over time, the number of spermatocytes and the levels of the meiotic regulators and Hsp90 interactors Hsp70-2, NASP and Cdc2 are reduced. We speculate that Hsp90α may be required to maintain and to activate these regulators and/or to disassemble the synaptonemal complex that holds homologous chromosomes together. The link between fertility and Hsp90 is further supported by our finding that an Hsp90 inhibitor that can cross the blood-testis barrier can partially phenocopy the genetic defects.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Fase Paquiteno , Espermatocitos/citología , Testículo/metabolismo , Animales , Barrera Hematotesticular , Progresión de la Enfermedad , Femenino , Masculino , Meiosis , Ratones , Mutación , Fenotipo , Espermatogénesis , Espermatozoides/fisiología
19.
EMBO J ; 28(17): 2616-24, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19644444

RESUMEN

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.


Asunto(s)
Histonas/metabolismo , Meiosis/genética , Recombinación Genética , Animales , Intercambio Genético , Roturas del ADN de Doble Cadena , Reparación del ADN , Ratones , Ratones Endogámicos
20.
PLoS Genet ; 5(2): e1000393, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19247432

RESUMEN

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.


Asunto(s)
Emparejamiento Cromosómico , Reparación del ADN , Proteínas Nucleares/genética , Complejo Sinaptonémico/metabolismo , Animales , Femenino , Gametogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Unión Proteica , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Recombinación Genética , Espermatocitos/citología , Espermatocitos/metabolismo , Complejo Sinaptonémico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA