Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 136(3): 420-34, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19203578

RESUMEN

The biological response to DNA double-strand breaks acts to preserve genome integrity. Individuals bearing inactivating mutations in components of this response exhibit clinical symptoms that include cellular radiosensitivity, immunodeficiency, and cancer predisposition. The archetype for such disorders is Ataxia-Telangiectasia caused by biallelic mutation in ATM, a central component of the DNA damage response. Here, we report that the ubiquitin ligase RNF168 is mutated in the RIDDLE syndrome, a recently discovered immunodeficiency and radiosensitivity disorder. We show that RNF168 is recruited to sites of DNA damage by binding to ubiquitylated histone H2A. RNF168 acts with UBC13 to amplify the RNF8-dependent histone ubiquitylation by targeting H2A-type histones and by promoting the formation of lysine 63-linked ubiquitin conjugates. These RNF168-dependent chromatin modifications orchestrate the accumulation of 53BP1 and BRCA1 to DNA lesions, and their loss is the likely cause of the cellular and developmental phenotypes associated with RIDDLE syndrome.


Asunto(s)
Daño del ADN , Síndromes de Inmunodeficiencia/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Línea Celular , Histonas/metabolismo , Humanos , Síndromes de Inmunodeficiencia/genética , Tolerancia a Radiación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Nucleic Acids Res ; 45(D1): D369-D379, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-27980099

RESUMEN

The Biological General Repository for Interaction Datasets (BioGRID: https://thebiogrid.org) is an open access database dedicated to the annotation and archival of protein, genetic and chemical interactions for all major model organism species and humans. As of September 2016 (build 3.4.140), the BioGRID contains 1 072 173 genetic and protein interactions, and 38 559 post-translational modifications, as manually annotated from 48 114 publications. This dataset represents interaction records for 66 model organisms and represents a 30% increase compared to the previous 2015 BioGRID update. BioGRID curates the biomedical literature for major model organism species, including humans, with a recent emphasis on central biological processes and specific human diseases. To facilitate network-based approaches to drug discovery, BioGRID now incorporates 27 501 chemical-protein interactions for human drug targets, as drawn from the DrugBank database. A new dynamic interaction network viewer allows the easy navigation and filtering of all genetic and protein interaction data, as well as for bioactive compounds and their established targets. BioGRID data are directly downloadable without restriction in a variety of standardized formats and are freely distributed through partner model organism databases and meta-databases.


Asunto(s)
Biología Computacional , Bases de Datos Genéticas , Proteínas , Animales , Biología Computacional/métodos , Curaduría de Datos , Minería de Datos , Humanos , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Programas Informáticos
3.
Mol Cell ; 40(4): 619-31, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21055983

RESUMEN

Genome integrity is jeopardized each time DNA replication forks stall or collapse. Here we report the identification of a complex composed of MMS22L (C6ORF167) and TONSL (NFKBIL2) that participates in the recovery from replication stress. MMS22L and TONSL are homologous to yeast Mms22 and plant Tonsoku/Brushy1, respectively. MMS22L-TONSL accumulates at regions of ssDNA associated with distressed replication forks or at processed DNA breaks, and its depletion results in high levels of endogenous DNA double-strand breaks caused by an inability to complete DNA synthesis after replication fork collapse. Moreover, cells depleted of MMS22L are highly sensitive to camptothecin, a topoisomerase I poison that impairs DNA replication progression. Finally, MMS22L and TONSL are necessary for the efficient formation of RAD51 foci after DNA damage, and their depletion impairs homologous recombination. These results indicate that MMS22L and TONSL are genome caretakers that stimulate the recombination-dependent repair of stalled or collapsed replication forks.


Asunto(s)
Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Recombinación Genética , Estrés Fisiológico , Supervivencia Celular , Roturas del ADN de Doble Cadena , Células HeLa , Humanos , FN-kappa B/química , Unión Proteica , Fase S , Moldes Genéticos
4.
J Cell Biol ; 171(3): 447-58, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16260499

RESUMEN

Mammalian MutL homologues function in DNA mismatch repair (MMR) after replication errors and in meiotic recombination. Both functions are initiated by a heterodimer of MutS homologues specific to either MMR (MSH2-MSH3 or MSH2-MSH6) or crossing over (MSH4-MSH5). Mutations of three of the four MutL homologues (Mlh1, Mlh3, and Pms2) result in meiotic defects. We show herein that two distinct complexes involving MLH3 are formed during murine meiosis. The first is a stable association between MLH3 and MLH1 and is involved in promoting crossing over in conjunction with MSH4-MSH5. The second complex involves MLH3 together with MSH2-MSH3 and localizes to repetitive sequences at centromeres and the Y chromosome. This complex is up-regulated in Pms2-/- males, but not females, providing an explanation for the sexual dimorphism seen in Pms2-/- mice. The association of MLH3 with repetitive DNA sequences is coincident with MSH2-MSH3 and is decreased in Msh2-/- and Msh3-/- mice, suggesting a novel role for the MMR family in the maintenance of repeat unit integrity during mammalian meiosis.


Asunto(s)
Proteínas Portadoras/metabolismo , Profase Meiótica I , Proteína 2 Homóloga a MutS/metabolismo , Cromosoma Y/fisiología , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/metabolismo , Centrómero/genética , Centrómero/fisiología , Centrómero/ultraestructura , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Masculino , Meiosis , Ratones , Ratones Noqueados , Microscopía Inmunoelectrónica , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteína 2 Homóloga a MutS/genética , Proteína 3 Homóloga de MutS , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Espermatocitos/fisiología , Espermatocitos/ultraestructura , Cromosoma Y/genética , Cromosoma Y/ultraestructura
5.
Curr Biol ; 16(8): R296-9, 2006 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-16631579

RESUMEN

DNA polymerase zeta and Rev1 play key roles in replication past DNA lesions. New work shows that the yeast checkpoint kinase Mec1 recruits a complex consisting of polymerase zeta and Rev1 to DNA double-strand breaks. This study highlights the role of polymerases that mediate translesion synthesis in the response to DNA double-strand breaks.


Asunto(s)
Reparación del ADN/fisiología , ADN de Hongos/biosíntesis , Nucleotidiltransferasas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas
6.
Genes (Basel) ; 2(1): 260-79, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21931878

RESUMEN

Meiosis is a highly conserved process, which is stringently regulated in all organisms, from fungi through to humans. Two major events define meiosis in eukaryotes. The first is the pairing, or synapsis, of homologous chromosomes and the second is the exchange of genetic information in a process called meiotic recombination. Synapsis is mediated by the meiosis-specific synaptonemal complex structure in combination with the cohesins that tether sister chromatids together along chromosome arms through prophase I. Previously, we identified FKBP6 as a novel component of the mammalian synaptonemal complex. Further studies demonstrated an interaction between FKBP6 and the NIMA-related kinase-1, NEK1. To further investigate the role of NEK1 in mammalian meiosis, we have examined gametogenesis in the spontaneous mutant, Nek1kat2J. Homozygous mutant animals show decreased testis size, defects in testis morphology, and in cohesin removal at late prophase I of meiosis, causing complete male infertility. Cohesin protein SMC3 remains localized to the meiotic chromosome cores at diplonema in the Nek1 mutant, and also in the related Fkbp6 mutant, while in wild type cells SMC3 is removed from the cores at the end of prophase I and becomes more diffuse throughout the DAPI stained region of the nucleus. These data implicate NEK1 as a possible kinase involved in cohesin redistribution in murine spermatocytes.

7.
Biol Reprod ; 78(3): 462-71, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18057311

RESUMEN

The DNA mismatch repair (MMR) family functions in a variety of contexts to preserve genome integrity in most eukaryotes. In particular, members of the MMR family are involved in the process of meiotic recombination in germ cells. MMR gene mutations in mice result in meiotic disruption during prophase I, but the extent of this disruption often differs between male and female meiocytes. To address the role of MMR proteins specifically in female meiosis, we explored the progression of oocytes through prophase I and the meiotic divisions in mice harboring deletions in members of the MMR pathway (Mlh1, Mlh3, Exo1, and an ATPase-deficient variant of Mlh1, Mlh1(G67R)). The colocalization of MLH1 and MLH3, key proteins involved in stabilization of nascent crossovers, was dependent on intact heterodimer formation and was highly correlated with the ability of oocytes to progress through to metaphase II. The exception was Exo1(-/-) oocytes, in which normal MLH1/MLH3 localization was observed followed by failure to proceed to metaphase II. All mutant oocytes were able to resume meiosis after dictyate arrest, but they showed a dramatic decline in chiasmata (to less than 25% of normal), accompanied by varied progression through metaphase I. Taken together, these results demonstrate that MMR function is required for the formation and stabilization of crossovers in mammalian oocytes and that, in the absence of a functional MMR system, the failure to maintain chiasmata results in a reduced ability to proceed normally through the first and second meiotic divisions, despite near-normal levels of meiotic resumption after dictyate arrest.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Mutación de Línea Germinal , Meiosis/genética , Meiosis/fisiología , Preñez , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Exodesoxirribonucleasas/genética , Femenino , Frecuencia de los Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/metabolismo , Embarazo , Transducción de Señal/genética
8.
Science ; 318(5856): 1637-40, 2007 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-18006705

RESUMEN

Cells respond to DNA double-strand breaks by recruiting factors such as the DNA-damage mediator protein MDC1, the p53-binding protein 1 (53BP1), and the breast cancer susceptibility protein BRCA1 to sites of damaged DNA. Here, we reveal that the ubiquitin ligase RNF8 mediates ubiquitin conjugation and 53BP1 and BRCA1 focal accumulation at sites of DNA lesions. Moreover, we establish that MDC1 recruits RNF8 through phosphodependent interactions between the RNF8 forkhead-associated domain and motifs in MDC1 that are phosphorylated by the DNA-damage activated protein kinase ataxia telangiectasia mutated (ATM). We also show that depletion of the E2 enzyme UBC13 impairs 53BP1 recruitment to sites of damage, which suggests that it cooperates with RNF8. Finally, we reveal that RNF8 promotes the G2/M DNA damage checkpoint and resistance to ionizing radiation. These results demonstrate how the DNA-damage response is orchestrated by ATM-dependent phosphorylation of MDC1 and RNF8-mediated ubiquitination.


Asunto(s)
Estructuras del Núcleo Celular/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Reparación del ADN , Proteínas de Unión al ADN/química , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , ARN Interferente Pequeño , Transactivadores/química , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación
9.
Chromosoma ; 114(2): 92-102, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15983832

RESUMEN

Genetic modifications causing germ cell death during meiotic prophase in the mouse frequently have sexually dimorphic phenotypes where oocytes reach more advanced stages than spermatocytes. To determine to what extent these dimorphisms are due to differences in male versus female meiotic prophase development, we compared meiotic chromosome events in the two sexes in both wild-type and mutant mice. We report the abundance and time course of appearance of structural and recombination-related proteins of fetal oocyte nuclei. Oocytes at successive days post coitus show rapid, synchronous meiotic prophase development compared with the continuous spermatocyte development in adult testis. Consequently, a genetic defect requiring 2-3 days from the onset of prophase to reach arrest registers pachytene as the developmental endpoint in oocytes. Pachytene spermatocytes, on the other hand, which normally accumulate during days 4-10 after the onset of prophase, will be rare, giving the appearance of an earlier endpoint than in oocytes. We conclude that these different logistics create apparent sexually dimorphic endpoints. For more pronounced sexual dimorphisms, we examined meiotic prophase of mice with genetic modifications of meiotic chromosome core components that cause male but not female sterility. The correlations between male sterility and alterations in the organization of the sex chromosome cores and X-Y chromatin may indicate that impaired signals from the XY domain (XY chromosome cores, chromatin, dense body and sex body) may interfere with the progression of the spermatocyte through prophase. Oocytes, in the absence of the X-Y pair, do not suffer such defects.


Asunto(s)
Infertilidad Masculina/genética , Espermatocitos/fisiología , Cromosoma X/genética , Cromosoma Y/genética , Animales , Proteína BRCA1/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Oocitos/fisiología
10.
J Cell Sci ; 115(Pt 8): 1611-22, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11950880

RESUMEN

During mouse meiosis, the early prophase RAD51/DMC1 recombination protein sites, which are associated with the chromosome cores and which serve as markers for ongoing DNA-DNA interactions, are in ten-fold excess of the eventual reciprocal recombinant events. Most, if not all, of these early interactions are eliminated as prophase progresses. The manner in which these sites are eliminated is the focus of this investigation. We report that these sites acquire replication protein A, RPA and the Escherichia coli MUTS homologue, MSH4p, and somewhat later the Bloom helicase, BLM, while simultaneously losing the RAD51/DMC1 component. Eventually the RPA component is also lost and BLM sites remain. At that time, the MUTL homologue, MLH1p, which is essential for reciprocal recombination in the mouse, appears in numbers and locations that correspond to the distribution of reciprocal recombination events. However, the MLH1 foci do not appear to coincide with the remaining BLM sites. The MLH1p is specifically localized to electron-microscope-defined recombination nodules. We consider the possibility that the homology-search RAD51/DMC1 complexes are involved in homologous chromosome synapsis but that most of these early DNA-DNA interactions are later resolved by the anti-recombination RPA/MSH4/BLM-topoisomerase complex, thereby preventing the formation of superfluous reciprocal recombinant events.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , ADN/metabolismo , Meiosis/fisiología , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/metabolismo , Animales , Cromosomas/ultraestructura , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Masculino , Ratones , Microscopía Inmunoelectrónica , Modelos Genéticos , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/metabolismo , Recombinasa Rad51 , Ratas , RecQ Helicasas , Espermatocitos/citología , Factores de Tiempo
11.
Genes Dev ; 17(5): 603-14, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12629043

RESUMEN

Exonuclease 1 (Exo1) is a 5'-3' exonuclease that interacts with MutS and MutL homologs and has been implicated in the excision step of DNA mismatch repair. To investigate the role of Exo1 in mammalian mismatch repair and assess its importance for tumorigenesis and meiosis, we generated an Exo1 mutant mouse line. Analysis of Exo1(-/-) cells for mismatch repair activity in vitro showed that Exo1 is required for the repair of base:base and single-base insertion/deletion mismatches in both 5' and 3' nick-directed repair. The repair defect in Exo1(-/-) cells also caused elevated microsatellite instability at a mononucleotide repeat marker and a significant increase in mutation rate at the Hprt locus. Exo1(-/-) animals displayed reduced survival and increased susceptibility to the development of lymphomas. In addition, Exo1(-/-) male and female mice were sterile because of a meiotic defect. Meiosis in Exo1(-/-) animals proceeded through prophase I; however, the chromosomes exhibited dynamic loss of chiasmata during metaphase I, resulting in meiotic failure and apoptosis. Our results show that mammalian Exo1 functions in mutation avoidance and is essential for male and female meiosis.


Asunto(s)
Reparación del ADN/fisiología , Exodesoxirribonucleasas/metabolismo , Predisposición Genética a la Enfermedad , Infertilidad/genética , Neoplasias/genética , Animales , Disparidad de Par Base/genética , Blastocisto , Línea Celular , Reparación del ADN/genética , Exodesoxirribonucleasas/genética , Femenino , Marcación de Gen , Infertilidad/etiología , Masculino , Meiosis/fisiología , Metafase/fisiología , Ratones/embriología , Repeticiones de Microsatélite
12.
Science ; 300(5623): 1291-5, 2003 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-12764197

RESUMEN

Meiosis is a critical stage of gametogenesis in which alignment and synapsis of chromosomal pairs occur, allowing for the recombination of maternal and paternal genomes. Here we show that FK506 binding protein (Fkbp6) localizes to meiotic chromosome cores and regions of homologous chromosome synapsis. Targeted inactivation of Fkbp6 in mice results in aspermic males and the absence of normal pachytene spermatocytes. Moreover, we identified the deletion of Fkbp6 exon 8 as the causative mutation in spontaneously male sterile as/as mutant rats. Loss of Fkbp6 results in abnormal pairing and misalignments between homologous chromosomes, nonhomologous partner switches, and autosynapsis of X chromosome cores in meiotic spermatocytes. Fertility and meiosis are normal in Fkbp6 mutant females. Thus, Fkbp6 is a component of the synaptonemal complex essential for sex-specific fertility and for the fidelity of homologous chromosome pairing in meiosis.


Asunto(s)
Emparejamiento Cromosómico/fisiología , Fertilidad/fisiología , Infertilidad Masculina/fisiopatología , Meiosis , Complejo Sinaptonémico/fisiología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Proteínas de Ciclo Celular , Clonación Molecular , Proteínas de Unión al ADN , Exones , Femenino , Marcación de Gen , Humanos , Infertilidad Masculina/genética , Masculino , Ratones , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oogénesis , Ovario/fisiología , Profase , Ratas , Eliminación de Secuencia , Espermátides/fisiología , Espermatocitos/fisiología , Espermatocitos/ultraestructura , Espermatogénesis , Proteínas de Unión a Tacrolimus/química , Testículo/fisiología , Cromosoma X/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA