Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
G3 (Bethesda) ; 14(9)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-38985658

RESUMEN

One of the major functions of programmed cell death (apoptosis) is the removal of cells that suffered oncogenic mutations, thereby preventing cancerous transformation. By making use of a Double-Headed-EP (DEP) transposon, a P element derivative made in our laboratory, we made an insertional mutagenesis screen in Drosophila melanogaster to identify genes that, when overexpressed, suppress the p53-activated apoptosis. The DEP element has Gal4-activatable, outward-directed UAS promoters at both ends, which can be deleted separately in vivo. In the DEP insertion mutants, we used the GMR-Gal4 driver to induce transcription from both UAS promoters and tested the suppression effect on the apoptotic rough eye phenotype generated by an activated UAS-p53 transgene. By DEP insertions, 7 genes were identified, which suppressed the p53-induced apoptosis. In 4 mutants, the suppression effect resulted from single genes activated by 1 UAS promoter (Pka-R2, Rga, crol, and Spt5). In the other 3 (Orct2, Polr2M, and stg), deleting either UAS promoter eliminated the suppression effect. In qPCR experiments, we found that the genes in the vicinity of the DEP insertion also showed an elevated expression level. This suggested an additive effect of the nearby genes on suppressing apoptosis. In the eukaryotic genomes, there are coexpressed gene clusters. Three of the DEP insertion mutants are included, and 2 are in close vicinity of separate coexpressed gene clusters. This raises the possibility that the activity of some of the genes in these clusters may help the suppression of the apoptotic cell death.


Asunto(s)
Apoptosis , Proteínas de Drosophila , Drosophila melanogaster , Mutagénesis Insercional , Proteína p53 Supresora de Tumor , Animales , Elementos Transponibles de ADN , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genes Dominantes , Genes Supresores , Mutagénesis Insercional/métodos , Fenotipo , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Nature ; 518(7538): 258-62, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25642963

RESUMEN

Large-scale genomic studies have shown that half of epithelial ovarian cancers (EOCs) have alterations in genes regulating homologous recombination (HR) repair. Loss of HR accounts for the genomic instability of EOCs and for their cellular hyper-dependence on alternative poly-ADP ribose polymerase (PARP)-mediated DNA repair mechanisms. Previous studies have implicated the DNA polymerase θ (Polθ also known as POLQ, encoded by POLQ) in a pathway required for the repair of DNA double-strand breaks, referred to as the error-prone microhomology-mediated end-joining (MMEJ) pathway. Whether Polθ interacts with canonical DNA repair pathways to prevent genomic instability remains unknown. Here we report an inverse correlation between HR activity and Polθ expression in EOCs. Knockdown of Polθ in HR-proficient cells upregulates HR activity and RAD51 nucleofilament assembly, while knockdown of Polθ in HR-deficient EOCs enhances cell death. Consistent with these results, genetic inactivation of an HR gene (Fancd2) and Polq in mice results in embryonic lethality. Moreover, Polθ contains RAD51 binding motifs and it blocks RAD51-mediated recombination. Our results reveal a synthetic lethal relationship between the HR pathway and Polθ-mediated repair in EOCs, and identify Polθ as a novel druggable target for cancer therapy.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Polimerasa Dirigida por ADN/metabolismo , Recombinación Homóloga , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Secuencias de Aminoácidos , Animales , Carcinoma Epitelial de Ovario , Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades/genética , Replicación del ADN , ADN Polimerasa Dirigida por ADN/deficiencia , Pérdida del Embrión , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Femenino , Inestabilidad Genómica , Recombinación Homóloga/genética , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Unión Proteica , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación/genética , ADN Polimerasa theta
3.
Mol Cell ; 47(3): 396-409, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22704558

RESUMEN

Completion of DNA replication after replication stress depends on PCNA, which undergoes monoubiquitination to stimulate direct bypass of DNA lesions by specialized DNA polymerases or is polyubiquitinated to promote recombination-dependent DNA synthesis across DNA lesions by template switching mechanisms. Here we report that the ZRANB3 translocase, a SNF2 family member related to the SIOD disorder SMARCAL1 protein, is recruited by polyubiquitinated PCNA to promote fork restart following replication arrest. ZRANB3 depletion in mammalian cells results in an increased frequency of sister chromatid exchange and DNA damage sensitivity after treatment with agents that cause replication stress. Using in vitro biochemical assays, we show that recombinant ZRANB3 remodels DNA structures mimicking stalled replication forks and disassembles recombination intermediates. We therefore propose that ZRANB3 maintains genomic stability at stalled or collapsed replication forks by facilitating fork restart and limiting inappropriate recombination that could occur during template switching events.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Poliubiquitina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estrés Fisiológico/genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Daño del ADN/fisiología , ADN Helicasas/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Datos de Secuencia Molecular , Osteosarcoma , Unión Proteica/fisiología , Recombinación Genética/fisiología , Intercambio de Cromátides Hermanas/fisiología , Ubiquitinación/fisiología
4.
Proc Natl Acad Sci U S A ; 108(32): 13130-4, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788515

RESUMEN

Wolf-Hirschhorn syndrome (WHS) is a malformation syndrome associated with growth retardation, mental retardation, and immunodeficiency resulting from a hemizygous deletion of the short arm of chromosome 4, called the WHS critical region (WHSC). The WHSC1 gene is located in this region, and its loss is believed to be responsible for a number of WHS characteristics. We identified WHSC1 in a genetic screen for genes involved in responding to replication stress, linking Wolf-Hirschhorn syndrome to the DNA damage response (DDR). Here, we report that the WHSC1 protein is a member of the DDR pathway. WHSC1 localizes to sites of DNA damage and replication stress and is required for resistance to many DNA-damaging and replication stress-inducing agents. Through its SET domain, WHSC1 regulates the methylation status of the histone H4 K20 residue and is required for the recruitment of 53BP1 to sites of DNA damage. We propose that Wolf-Hirschhorn syndrome results from a defect in the DDR.


Asunto(s)
Daño del ADN , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas Represoras/metabolismo , Síndrome de Wolf-Hirschhorn/metabolismo , Síndrome de Wolf-Hirschhorn/patología , Línea Celular Tumoral , Quinasa de Punto de Control 2 , Replicación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/deficiencia , Histonas/metabolismo , Humanos , Hidroxiurea/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/metabolismo , Metilación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteína de Replicación A/metabolismo , Proteínas Represoras/deficiencia , Estrés Fisiológico/efectos de los fármacos , Proteína 1 de Unión al Supresor Tumoral P53
5.
Mol Cell Biol ; 30(3): 684-93, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19948885

RESUMEN

Unrepaired DNA lesions can block the progression of the replication fork, leading to genomic instability and cancer in higher-order eukaryotes. In Saccharomyces cerevisiae, replication through DNA lesions can be mediated by translesion synthesis DNA polymerases, leading to error-free or error-prone damage bypass, or by Rad5-mediated template switching to the sister chromatid that is inherently error free. While translesion synthesis pathways are highly conserved from yeast to humans, very little is known of a Rad5-like pathway in human cells. Here we show that a human homologue of Rad5, HLTF, can facilitate fork regression and has a role in replication of damaged DNA. We found that HLTF is able to reverse model replication forks, a process which depends on its double-stranded DNA translocase activity. Furthermore, from analysis of isolated dually labeled chromosomal fibers, we demonstrate that in vivo, HLTF promotes the restart of replication forks blocked at DNA lesions. These findings suggest that HLTF can promote error-free replication of damaged DNA and support a role for HLTF in preventing mutagenesis and carcinogenesis, providing thereby for its potential tumor suppressor role.


Asunto(s)
Daño del ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Interferencia de ARN , Factores de Transcripción/genética
6.
Proc Natl Acad Sci U S A ; 105(10): 3768-73, 2008 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-18316726

RESUMEN

Human helicase-like transcription factor (HLTF) is frequently inactivated in colorectal and gastric cancers. Here, we show that HLTF is a functional homologue of yeast Rad5 that promotes error-free replication through DNA lesions. HLTF and Rad5 share the same unique structural features, including a RING domain embedded within a SWI/SNF helicase domain and an HIRAN domain. We find that inactivation of HLTF renders human cells sensitive to UV and other DNA-damaging agents and that HLTF complements the UV sensitivity of a rad5Delta yeast strain. Also, similar to Rad5, HLTF physically interacts with the Rad6-Rad18 and Mms2-Ubc13 ubiquitin-conjugating enzyme complexes and promotes the Lys-63-linked polyubiquitination of proliferating cell nuclear antigen at its Lys-164 residue. A requirement of HLTF for error-free postreplication repair of damaged DNA is in keeping with its cancer-suppression role.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Poliubiquitina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adenosina Trifosfatasas/metabolismo , Línea Celular , ADN Helicasas , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/efectos de la radiación , Prueba de Complementación Genética , Humanos , Ligasas/metabolismo , Lisina/metabolismo , Metilmetanosulfonato/farmacología , Mutación/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/efectos de la radiación , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de la radiación , Proteínas de Saccharomyces cerevisiae/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinación/efectos de la radiación , Rayos Ultravioleta
7.
Proc Natl Acad Sci U S A ; 103(48): 18107-12, 2006 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17108083

RESUMEN

Human SHPRH gene is located at the 6q24 chromosomal region, and loss of heterozygosity in this region is seen in a wide variety of cancers. SHPRH is a member of the SWI/SNF family of ATPases/helicases, and it possesses a C(3)HC(4) RING motif characteristic of ubiquitin ligase proteins. In both of these features, SHPRH resembles the yeast Rad5 protein, which, together with Mms2-Ubc13, promotes replication through DNA lesions via an error-free postreplicational repair pathway. Genetic evidence in yeast has indicated a role for Rad5 as a ubiquitin ligase in mediating the Mms2-Ubc13-dependent polyubiquitylation of proliferating cell nuclear antigen. Here we show that SHPRH is a functional homolog of Rad5. Similar to Rad5, SHPRH physically interacts with the Rad6-Rad18 and Mms2-Ubc13 complexes, and we show that SHPRH protein is a ubiquitin ligase indispensable for Mms2-Ubc13-dependent polyubiquitylation of proliferating cell nuclear antigen. Based on these observations, we predict a role for SHPRH in promoting error-free replication through DNA lesions. Such a role for SHPRH is consistent with the observation that this gene is mutated in a number of cancer cell lines, including those from melanomas and ovarian cancers, which raises the strong possibility that SHPRH function is an important deterrent to mutagenesis and carcinogenesis in humans.


Asunto(s)
ADN Helicasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Línea Celular , ADN/genética , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/aislamiento & purificación , Humanos , Datos de Secuencia Molecular , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA