Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Cell ; 58(3): 468-82, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25921070

RESUMEN

Transcription is repressed if a DNA double-strand break (DSB) is introduced in close proximity to a transcriptional activation site at least in part by H2A-ubiquitination. While ATM signaling is involved, how it controls H2A-ubiquitination remains unclear. Here, we identify that, in response to DSBs, a transcriptional elongation factor, ENL (MLLT1), is phosphorylated by ATM at conserved SQ sites. This phosphorylation increases the interaction between ENL and the E3-ubiquitin-ligase complex of Polycomb Repressive Complex 1 (PRC1) via BMI1. This interaction promotes enrichment of PRC1 at transcription elongation sites near DSBs to ubiquitinate H2A leading to transcriptional repression. ENL SQ sites and BMI1 are necessary for KU70 accumulation at DSBs near active transcription sites and cellular resistance to DSBs. Our data suggest that ATM-dependent phosphorylation of ENL functions as switch from elongation to Polycomb-mediated repression to preserve genome integrity.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Reparación del ADN , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Western Blotting , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fosforilación , Complejo Represivo Polycomb 1/genética , Unión Proteica , Interferencia de ARN , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Transcripción Genética
2.
Cancer Sci ; 113(12): 4230-4243, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36082621

RESUMEN

Breast cancer gene 1 (BRCA1) plays roles in DNA repair and centrosome regulation and is involved in DNA damage-induced centrosome amplification (DDICA). Here, the centrosomal localization of BRCA1 and the kinases involved in centrosome duplication were analyzed in each cell cycle phase after treatment with DNA crosslinker cisplatin (CDDP). CDDP treatment increased the centrosomal localization of BRCA1 in early S-G2 phase. BRCA1 contributed to the increased centrosomal localization of Aurora A in S phase and that of phosphorylated Polo-like kinase 1 (PLK1) in late S phase after CDDP treatment, resulting in centriole disengagement and overduplication. The increased centrosomal localization of BRCA1 and Aurora A induced by CDDP treatment involved the nuclear export of BRCA1 and BRCA1 phosphorylation by ataxia telangiectasia mutated (ATM). Patient-derived variants and mutations at phosphorylated residues of BRCA1 suppressed the interaction between BRCA1 and Aurora A, as well as the CDDP-induced increase in the centrosomal localization of BRCA1 and Aurora A. These results suggest that CDDP induces the phosphorylation of BRCA1 by ATM in the nucleus and its transport to the cytoplasm, thereby promoting the centrosomal localization Aurora A, which phosphorylates PLK1. The function of BRCA1 in the translocation of the DNA damage signal from the nucleus to the centrosome to induce centrosome amplification after CDDP treatment might support its role as a tumor suppressor.


Asunto(s)
Aurora Quinasa A , Proteína BRCA1 , Centrosoma , Daño del ADN , Humanos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Fase G2 , Fosforilación , Aurora Quinasa A/metabolismo
3.
Genes Cells ; 26(5): 328-335, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33624391

RESUMEN

SIRT2 and SIRT3 protein deacetylases maintain genome integrity and stability. However, their mechanisms for maintaining the genome remain unclear. To examine the roles of SIRT2 and SIRT3 in DSB repair, I-SceI-based GFP reporter assays for HR, single-strand annealing (SSA) and nonhomologous end joining (NHEJ) repair were performed under SIRT2- or SIRT3-depleted conditions. SIRT2 or SIRT3 depletion inhibited HR repair equally to RAD52 depletion, but did not affect SSA and NHEJ repairs. SIRT2 or SIRT3 depletion disturbed the recruitment of RAD51 to DSB sites, an essential step for RAD51-dependent HR repair, but not directly through RAD52 deacetylation. SIRT2 or SIRT3 depletion decreased the colocalization of γH2AX foci with RPA1, and thus, they might be involved in initiating DSB end resection for the recruitment of RAD51 to DSB sites at an early step in HR repair. These results show the novel underlying mechanism of the SIRT2 and SIRT3 functions in HR for genome stability.


Asunto(s)
Recombinación Homóloga/genética , Reparación del ADN por Recombinación , Sirtuina 2/metabolismo , Sirtuina 3/metabolismo , Acetilación , Roturas del ADN de Doble Cadena , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Recombinasa Rad51/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo
4.
Mol Cell ; 53(4): 617-30, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24560272

RESUMEN

DNA double-strand breaks (DSBs) are deleterious lesions that lead to genetic mutations and cell death. Protein ubiquitination mediated by the E3 ubiquitin ligase RNF8 within the regions surrounding DSBs recruits DNA DSB response (DDR) factors and induces chromatin remodeling, which supports cell survival after DNA damage. Nevertheless, the impact of RNF8-mediated ubiquitination on DNA repair remains to be elucidated. Here, we report that depletion of the deubiquitinating enzyme OTUB2 enhances RNF8-mediated ubiquitination in an early phase of the DDR and promotes faster DSB repair but suppresses homologous recombination. The rapid ubiquitination results in accelerated accumulation of 53BP1 and RAP80 at DSBs, which in turn protects DSB ends from resection in OTUB2-depleted cells. Mechanistically, OTUB2 suppresses RNF8-mediated L3MBTL1 ubiquitination and Lys 63-linked ubiquitin chain formation in a deubiquitinating activity-dependent manner. Thus, OTUB2 fine-tunes the speed of DSB-induced ubiquitination so that the appropriate DNA repair pathway is chosen.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Tioléster Hidrolasas/química , Proteínas Portadoras/metabolismo , Muerte Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/química , Biblioteca de Genes , Silenciador del Gen , Células HeLa , Chaperonas de Histonas , Histonas/química , Recombinación Homóloga , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/química , Mutación , Proteínas Nucleares/metabolismo , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Recombinación Genética , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/química , Ubiquitina-Proteína Ligasas
5.
PLoS Genet ; 14(3): e1007277, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29590107

RESUMEN

The p300 and CBP histone acetyltransferases are recruited to DNA double-strand break (DSB) sites where they induce histone acetylation, thereby influencing the chromatin structure and DNA repair process. Whether p300/CBP at DSB sites also acetylate non-histone proteins, and how their acetylation affects DSB repair, remain unknown. Here we show that p300/CBP acetylate RAD52, a human homologous recombination (HR) DNA repair protein, at DSB sites. Using in vitro acetylated RAD52, we identified 13 potential acetylation sites in RAD52 by a mass spectrometry analysis. An immunofluorescence microscopy analysis revealed that RAD52 acetylation at DSBs sites is counteracted by SIRT2- and SIRT3-mediated deacetylation, and that non-acetylated RAD52 initially accumulates at DSB sites, but dissociates prematurely from them. In the absence of RAD52 acetylation, RAD51, which plays a central role in HR, also dissociates prematurely from DSB sites, and hence HR is impaired. Furthermore, inhibition of ataxia telangiectasia mutated (ATM) protein by siRNA or inhibitor treatment demonstrated that the acetylation of RAD52 at DSB sites is dependent on the ATM protein kinase activity, through the formation of RAD52, p300/CBP, SIRT2, and SIRT3 foci at DSB sites. Our findings clarify the importance of RAD52 acetylation in HR and its underlying mechanism.


Asunto(s)
Roturas del ADN de Doble Cadena , Histona Acetiltransferasas/fisiología , Histona Desacetilasas/fisiología , Recombinación Homóloga , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Acetilación , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Histona Acetiltransferasas/genética , Histona Desacetilasas/genética , Humanos , Microscopía Fluorescente , Técnicas del Sistema de Dos Híbridos
6.
Cancer Sci ; 111(5): 1443-1451, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32232911

RESUMEN

DNA double-strand break (DSB) is a serious type of DNA damage and is known to trigger multiple responses within cells. In these responses, novel relationships among DSB, DSB repair, and transcription machineries are created. First, transcription is repressed if DSB occurs near or at the transcription site, termed DSB-induced transcriptional repression, which contributes to DSB repair with the aid of DNA damage-signaling pathways, ATM- or DNA-PKcs-signaling pathways. DSB-induced transcriptional repression is also regulated by transcriptional factors TLP1, NELF, and ENL, as well as chromatin remodeling and organizing factors ZMYND8, CDYL1, PBAF, and cohesin. Second, transcription and RNA promote DSB repair for genome integrity. Transcription factors such as LEDGF, SETD2, and transcriptionally active histone modification, H3K36, facilitate homologous recombination to overcome DSB. At transcriptional active sites, DNA:RNA hybrids, termed R-loops, which are formed by DSB, are processed by RAD52 and XPG leading to an activation of the homologous recombination pathway. Even in a transcriptionally inactive non-genic sites, noncoding RNAs that are produced by RNA polymerase II, DICER, and DROSHA, help to recruit DSB repair proteins at the DSB sites. Third, transcriptional activation itself, however, can induce DSB. Transcriptional activation often generates specific DNA structures such as R-loops and topoisomerase-induced DSBs, which cause genotoxic stress and may lead to genome instability and consequently to cancer. Thus, transcription and DSB repair machineries interact and cooperate to prevent genome instability and cancer.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Inestabilidad Genómica/genética , Neoplasias/genética , Transcripción Genética , Daño del ADN , Recombinación Homóloga , Humanos , Neoplasias/metabolismo , ARN , Activación Transcripcional
7.
Mol Cell ; 40(6): 976-87, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21172662

RESUMEN

DNA double-strand breaks (DSBs) are repaired via nonhomologous end-joining (NHEJ) or homologous recombination (HR), but cellular repair processes remain elusive. We show here that the ATP-dependent chromatin-remodeling factors, ACF1 and SNF2H, accumulate rapidly at DSBs and are required for DSB repair in human cells. If the expression of ACF1 or SNF2H is suppressed, cells become extremely sensitive to X-rays and chemical treatments producing DSBs, and DSBs remain unrepaired. ACF1 interacts directly with KU70 and is required for the accumulation of KU proteins at DSBs. The KU70/80 complex becomes physically more associated with the chromatin-remodeling factors of the CHRAC complex, which includes ACF1, SNF2H, CHRAC15, and CHRAC17, after treatments producing DSBs. Furthermore, the frequency of NHEJ as well as HR induced by DSBs in chromosomal DNA is significantly decreased in cells depleted of either of these factors. Thus, ACF1 and its complexes play important roles in DSBs repair.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Factores de Transcripción/metabolismo , Antígenos Nucleares/metabolismo , Células Cultivadas , ADN Polimerasa III/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Autoantígeno Ku , Nucleoproteínas/metabolismo , Rayos Ultravioleta
8.
Carcinogenesis ; 34(11): 2486-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23825154

RESUMEN

Inhibitors of poly(ADP-ribose) polymerase (PARP) are promising anticancer drugs, particularly for the treatment of tumors deficient in the DNA damage response (DDR). However, it is challenging to design effective therapeutic strategies for use of these compounds against cancers without DDR deficiencies. In this context, combination therapies in which PARP inhibitors are used alongside DDR inhibitors have elicited a great deal of interest. Curcumin, a component of turmeric (Curcuma longa), has been tested in clinical studies for its chemosensitizing potential; however, the mechanisms of chemosensitization by curcumin have not been fully elucidated. This study demonstrates that curcumin suppresses three major DDR pathways: non-homologous end joining (NHEJ), homologous recombination (HR) and the DNA damage checkpoint. Curcumin suppresses the histone acetylation at DNA double-strand break (DSB) sites by inhibiting histone acetyltransferase activity, thereby reducing recruitment of the key NHEJ factor KU70/KU80 to DSB sites. Curcumin also suppresses HR by reducing expression of the BRCA1 gene, which regulates HR, by impairing histone acetylation at the BRCA1 promoter. Curcumin also inhibits ataxia telangiectasia and Rad3-related protein (ATR) kinase (IC50 in vitro = 493 nM), resulting in impaired activation of ATR-CHK1 signaling, which is necessary for HR and the DNA damage checkpoint pathway. Thus, curcumin suppresses three DDR pathways by inhibiting histone acetyltransferases and ATR. Concordantly, curcumin sensitizes cancer cells to PARP inhibitors by enhancing apoptosis and mitotic catastrophe via inhibition of both the DNA damage checkpoint and DSB repair. Our results indicate that curcumin is a promising sensitizer for PARP inhibitor-based therapy.


Asunto(s)
Curcumina/farmacología , Daño del ADN/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Transducción de Señal/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/metabolismo , Western Blotting , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Radioisótopos de Cobalto , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Rayos gamma , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Recombinación Homóloga/efectos de la radiación , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Regiones Promotoras Genéticas , Proteínas Quinasas/metabolismo , Sialoglicoproteínas/antagonistas & inhibidores , Sialoglicoproteínas/metabolismo , Transducción de Señal/efectos de la radiación , Células Tumorales Cultivadas , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Factores de Transcripción p300-CBP/metabolismo
9.
Oncogene ; 41(19): 2706-2718, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35393543

RESUMEN

DNA double-strand break (DSB) repair-pathway choice regulated by 53BP1 and BRCA1 contributes to genome stability. 53BP1 cooperates with the REV7-Shieldin complex and inhibits DNA end resection to block homologous recombination (HR) and affects the sensitivity to inhibitors for poly (ADP-ribose) polymerases (PARPs) in BRCA1-deficient cells. Here, we show that a REV7 binding protein, CHAMP1 (chromosome alignment-maintaining phosphoprotein 1), has an opposite function of REV7 in DSB repair and promotes HR through DNA end resection together with POGZ (POGO transposable element with ZNF domain). CHAMP1 was recruited to laser-micro-irradiation-induced DSB sites and promotes HR, but not NHEJ. CHAMP1 depletion suppressed the recruitment of BRCA1, but not the recruitment of 53BP1, suggesting that CHAMP1 regulates DSB repair pathway in favor of HR. Depletion of either CHAMP1 or POGZ impaired the recruitment of phosphorylated RPA2 and CtIP (CtBP-interacting protein) at DSB sites, implying that CHAMP1, in complex with POGZ, promotes DNA end resection for HR. Furthermore, loss of CHAMP1 and POGZ restored the sensitivity to a PARP inhibitor in cells depleted of 53BP1 together with BRCA1. These data suggest that CHAMP1and POGZ counteract the inhibitory effect of 53BP1 on HR by promoting DNA end resection and affect the resistance to PARP inhibitors.


Asunto(s)
Roturas del ADN de Doble Cadena , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteína BRCA1/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Recombinación Homóloga , Humanos , Fosfoproteínas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Transposasas/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
10.
Cancer Res ; 82(20): 3751-3762, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36166639

RESUMEN

Distinguishing oncogenic mutations from variants of unknown significance (VUS) is critical for precision cancer medicine. Here, computational modeling of 71,756 RET variants for positive selection together with functional assays of 110 representative variants identified a three-dimensional cluster of VUSs carried by multiple human cancers that cause amino acid substitutions in the calmodulin-like motif (CaLM) of RET. Molecular dynamics simulations indicated that CaLM mutations decrease interactions between Ca2+ and its surrounding residues and induce conformational distortion of the RET cysteine-rich domain containing the CaLM. RET-CaLM mutations caused ligand-independent constitutive activation of RET kinase by homodimerization mediated by illegitimate disulfide bond formation. RET-CaLM mutants possessed oncogenic and tumorigenic activities that could be suppressed by tyrosine kinase inhibitors targeting RET. This study identifies calcium-binding ablating mutations as a novel type of oncogenic mutation of RET and indicates that in silico-driven annotation of VUSs of druggable oncogenes is a promising strategy to identify targetable driver mutations. SIGNIFICANCE: Comprehensive proteogenomic and in silico analyses of a vast number of VUSs identify a novel set of oncogenic and druggable mutations in the well-characterized RET oncogene.


Asunto(s)
Proteínas de Drosophila , Neoplasia Endocrina Múltiple Tipo 2a , Neoplasias , Calcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Carcinogénesis/genética , Cisteína/genética , Cisteína/metabolismo , Disulfuros/metabolismo , Proteínas de Drosophila/genética , Humanos , Ligandos , Neoplasia Endocrina Múltiple Tipo 2a/genética , Neoplasia Endocrina Múltiple Tipo 2a/metabolismo , Mutación , Neoplasias/genética , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ret/genética
11.
DNA Repair (Amst) ; 7(6): 879-89, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18396468

RESUMEN

The protein Rad52 is a key player in various types of homologous recombination and is essential to maintenance of genomic integrity. Although evidence indicates that Rad52 is modified by SUMO, the physiological relevance of this sumoylation remains unclear. Here, we identify the conditions under which Rad52 sumoylation is induced, and clarify the role of this modification in homologous recombination. Oligomerization of Rad52 was a prerequisite for sumoylation, and the modification occurred in the cell proceeding S phase being exposed to the DNA-damaging agent methyl methanesulfonate (MMS). Following exposure to MMS, sumoylated Rad52 accumulated in rad51 cells, but not in the recombination-related gene mutants, rad54, rad55, rad59, sgs1, or srs2. The accumulation of sumoylated Rad52 was suppressed in rad51 cells expressing Rad51-K191R, an ATPase-defective protein presumed to be recruited to ssDNA. Although the sumoylation defective mutant rad52-3KR (K10R/K11R/K220R) showed no defect in mating-type switching, which did not lead to Rad52 sumoylation in wild-type cells, the mutant did demonstrate a partial defect in MMS-induced interchromosomal homologous recombination.


Asunto(s)
Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Secuencia de Bases , ADN/efectos de los fármacos , Cartilla de ADN , Metilmetanosulfonato/farmacología , Mutagénesis Sitio-Dirigida , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteínas de Saccharomyces cerevisiae/genética
12.
Nucleic Acids Res ; 35(2): 353-62, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17170004

RESUMEN

The inaccurate repair of DNA double-strand breaks (DSBs) can result in genomic instability, and additionally cell death or the development of cancer. Elg1, which forms an alternative RFC-like complex with RFC2-5, is required for the maintenance of genome stability in Saccharomyces cerevisiae, and its function has been linked to DNA replication or damage checkpoint response. Here, we show that Elg1 is involved in homologous recombination (HR)-mediated DSB repair. Mutants of elg1 were partially defective in HR induced by methylmethanesufonate (MMS) and phleomycin. Deletion of ELG1 resulted in less efficient repair of phleomycin-induced DSBs in G2/M phase-arrested cells. During HR between MAT and HML loci, Elg1 associated with both the MAT locus near the HO endonuclease-induced DSB site, and the HML homologous donor locus. The association of Elg1 with the MAT locus was not dependent on Rad52. However, Elg1 association with the HML locus depended on Rad52. Importantly, we found that two of the later steps in HR-mediated repair of an HO endonuclease-induced DSB, primer extension after strand invasion and ligation, were less efficient in elg1 mutants. Our results suggest that Elg1 is involved in DSB repair by HR.


Asunto(s)
Proteínas Portadoras/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas Portadoras/genética , Ciclo Celular , Genes del Tipo Sexual de los Hongos , Mutación , Fleomicinas/toxicidad , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Recombinación Genética , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae
13.
Nucleic Acids Res ; 35(15): 4989-5000, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17636314

RESUMEN

The efficient repair of double-strand breaks (DSBs) is crucial in maintaining genomic integrity. Sister chromatid cohesion is important for not only faithful chromosome segregation but also for proper DSB repair. During DSB repair, the Smc1-Smc3 cohesin complex is loaded onto chromatin around the DSB to support recombination-mediated DSB repair. In this study, we investigated whether Ctf18, a factor implicated in the establishment of sister chromatid cohesion, is involved in DSB repair in budding yeast. Ctf18 was recruited to HO-endonuclease induced DSB sites in an Mre11-dependent manner and to damaged chromatin in G2/M phase-arrested cells. The ctf18 mutant cells showed high sensitivity to DSB-inducible genotoxic agents and defects in DSB repair, as well as defects in damage-induced recombination between sister chromatids and between homologous chromosomes. These results suggest that Ctf18 is involved in damage-induced homologous recombination.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona , Replicación del ADN , Endodesoxirribonucleasas/fisiología , Exodesoxirribonucleasas/fisiología , Eliminación de Gen , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Nucleic Acids Res ; 35(9): 3109-17, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17452364

RESUMEN

The actin-related proteins (Arps) comprise a conserved protein family. Arp4p is found in large multisubunits of the INO80 and SWR1 chromatin remodeling complexes and in the NuA4 histone acetyltransferase complex. Here we show that arp4 (arp4S23A/D159A) temperature-sensitive cells are defective in G2/M phase function. arp4 mutants are sensitive to the microtubule depolymerizing agent benomyl and arrest at G2/M phase at restrictive temperature. Arp4p is associated with centromeric and telomeric regions throughout cell cycle. Ino80p, Esa1p and Swr1p, components of the INO80, NuA4 and SWR1 complexes, respectively, also associate with centromeres. The association of many kinetochore components including Cse4p, a component of the centromere nucleosome, Mtw1p and Ctf3p is partially impaired in arp4 cells, suggesting that the G2/M arrest of arp4 mutant cells is due to a defect in formation of the chromosomal segregation apparatus.


Asunto(s)
Actinas/fisiología , Ciclo Celular , Cinetocoros/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Actinas/análisis , Actinas/genética , Sitios de Unión , Ciclo Celular/efectos de los fármacos , División Celular , Fase G2 , Genómica , Mutación , Nocodazol/farmacología , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/análisis , Proteínas de Saccharomyces cerevisiae/genética
15.
Nucleic Acids Res ; 34(11): 3389-98, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16840526

RESUMEN

Dpb11 is required for the loading of DNA polymerases alpha and epsilon on to DNA in chromosomal DNA replication and interacts with the DNA damage checkpoint protein Ddc1 in Saccharomyces cerevisiae. The interaction between the homologs of Dpb11 and Ddc1 in human cells and fission yeast is thought to reflect their involvement in the checkpoint response. Here we show that dpb11-1 cells, carrying a mutated Dpb11 that cannot interact with Ddc1, are defective in the repair of methyl methanesulfonate (MMS)-induced DNA damage but not in the DNA damage checkpoint at the permissive temperature. Epistatic analyses suggested that Dpb11 is involved in the Rad51/Rad52-dependent recombination pathway. Ddc1 as well as Dpb11 were required for homologous recombination induced by MMS. Moreover, we found the in vivo association of Dpb11 and Ddc1 with not only the HO-induced double-strand break (DSB) site at MAT locus but also the donor sequence HML during homologous recombination between MAT and HML. Rad51 was required for their association with the HML donor locus, but not with DSB site at the MAT locus. In addition, the association of Dpb11 with the MAT and HML locus after induction of HO-induced DSB was dependent on Ddc1. These results indicate that, besides the involvement in the replication and checkpoint, Dpb11 functions with Ddc1 in the recombination repair process itself.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Reparación del ADN , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/genética , Daño del ADN , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Metilmetanosulfonato/toxicidad , Mutación , Fosfoproteínas/fisiología , Recombinasa Rad51/fisiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
17.
Genes Genet Syst ; 82(2): 157-60, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17507781

RESUMEN

The structural maintenance of chromosomes (SMC) family proteins (Smc1-Smc6) typically consist of two coiled-coil domains, an amino-terminal head domain, and a carboxyl-terminal tail domain. Rad50, a component of the Mre11/Rad50/Xrs2 (MRX) complex, has a similar domain structure to the SMC proteins. In Saccharomyces cerevisiae, the MRX complex appears to be essential for recombination between homologous chromosomes in meiotic cells, but not in cells undergoing vegetative growth. Here we provide for the first time evidence that Rad50, like Smc6, is required for the induction of recombination between homologous chromosomes in cells in the vegetative growth state upon exposure to methyl methanesulfonate. However, UV-induced recombination between homologous chromosomes is intact in both rad50 and smc6-56 mutant cells.


Asunto(s)
Cromosomas Fúngicos , Proteínas de Unión al ADN/fisiología , Metilmetanosulfonato/farmacología , Mitosis/efectos de los fármacos , Recombinación Genética/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/fisiología , Homología de Secuencia , Proliferación Celular/efectos de los fármacos , Mitosis/genética , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética
18.
Mutat Res ; 617(1-2): 90-7, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17289091

RESUMEN

Ortho-phenyl phenol (OPP) is broad-spectrum of fungicides and antibacterial agents. OPP tested negative in an Ames system and positive with respect to the formation of tumors in the urinary bladder in rats when administered in diet, showing attributes of an Ames test-negative carcinogen. It has also been demonstrated that OPP does not bind or cleave DNA in vivo or in vitro, rather dose-dependent protein binding in OPP-treated rats was observed. OPP, however, generates chromosomal aberrations including aneuploidy. Thus, the steps by which Ames test-negative carcinogens exert their effects need to be elucidated. Here, we used an assay of loss of heterozygosity (LOH) in Saccharomyces cerevisiae to determine the biological effects of OPP and its hepatic metabolite phenyl hydroquinone (PHQ). LOH was found to be induced by OPP and PHQ because of a functional chromosome loss: aneuploidy. PHQ bound to and interfered with the depolymerization of tubulin in vitro and arrested the cell-cycle at M and G1. These results indicate that OPP and PHQ damaged tubulin to cause mis-segregation of chromosome by delaying cell-cycle progression through mitosis, and as a consequence caused aneuploidy.


Asunto(s)
Aneuploidia , Compuestos de Bifenilo/metabolismo , Carcinógenos/metabolismo , Hidroquinonas/metabolismo , Saccharomyces cerevisiae/genética , Tubulina (Proteína)/metabolismo , División Celular , Cromosomas Fúngicos/genética , Citometría de Flujo , Fase G1 , Pérdida de Heterocigocidad , Microtúbulos/metabolismo
19.
Philos Trans R Soc Lond B Biol Sci ; 372(1731)2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28847829

RESUMEN

Nucleosome remodelling (NR) regulates transcription in an ATP-dependent manner, and influences gene expression required for development and cellular functions, including those involved in anti-cancer and anti-ageing processes. ATP-utilizing chromatin assembly and remodelling factor (ACF) and Brahma-associated factor (BAF) complexes, belonging to the ISWI and SWI/SNF families, respectively, are involved in various types of DNA repair. Suppression of several BAF factors makes U2OS cells significantly sensitive to X-rays, UV and especially to cisplatin, and these BAF factors contribute to the accumulation of repair proteins at various types of DNA damage and to DNA repair. Recent cancer genome sequencing and expression analysis has shown that BAF factors are frequently mutated or, more frequently, silenced in various types of cancer cells. Thus, those cancer cells are potentially X-ray- and especially cisplatin-sensitive, suggesting a way of optimizing current cancer therapy. Recent single-stem cell analysis suggests that mutations and epigenetic changes influence stem cell functionality leading to cellular ageing. Genetic and epigenetic changes in the BAF factors diminish DNA repair as well as transcriptional regulation activities, and DNA repair defects in turn negatively influence NR and transcriptional regulation. Thus, they build negative feedback loops, which accelerate both cellular senescence and transformation as common and rare cellular events, respectively, causing cellular ageing.This article is part of the themed issue 'Chromatin modifiers and remodellers in DNA repair and signalling'.


Asunto(s)
Senescencia Celular , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Regulación de la Expresión Génica , Neoplasias/genética , Nucleosomas/genética , Reparación del ADN , Humanos , Nucleosomas/metabolismo
20.
Nat Commun ; 8: 16102, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28719581

RESUMEN

HBO1, a histone acetyl transferase, is a co-activator of DNA pre-replication complex formation. We recently reported that HBO1 is phosphorylated by ATM and/or ATR and binds to DDB2 after ultraviolet irradiation. Here, we show that phosphorylated HBO1 at cyclobutane pyrimidine dimer (CPD) sites mediates histone acetylation to facilitate recruitment of XPC at the damaged DNA sites. Furthermore, HBO1 facilitates accumulation of SNF2H and ACF1, an ATP-dependent chromatin remodelling complex, to CPD sites. Depletion of HBO1 inhibited repair of CPDs and sensitized cells to ultraviolet irradiation. However, depletion of HBO1 in cells derived from xeroderma pigmentosum patient complementation groups, XPE, XPC and XPA, did not lead to additional sensitivity towards ultraviolet irradiation. Our findings suggest that HBO1 acts in concert with SNF2H-ACF1 to make the chromosome structure more accessible to canonical nucleotide excision repair factors.


Asunto(s)
Reparación del ADN , Histona Acetiltransferasas/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Fosforilación , Dímeros de Pirimidina/metabolismo , Factores de Transcripción/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA