Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Sci ; 113(12): 4230-4243, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36082621

RESUMO

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.


Assuntos
Aurora Quinase A , Proteína BRCA1 , Centrossomo , Dano ao DNA , Humanos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Fase G2 , Fosforilação , Aurora Quinase A/metabolismo
2.
Cancer Res ; 82(20): 3751-3762, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36166639

RESUMO

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.


Assuntos
Proteínas de Drosophila , Neoplasia Endócrina Múltipla Tipo 2a , Neoplasias , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Carcinogênese/genética , Cisteína/genética , Cisteína/metabolismo , Dissulfetos/metabolismo , Proteínas de Drosophila/genética , Humanos , Ligantes , Neoplasia Endócrina Múltipla Tipo 2a/genética , Neoplasia Endócrina Múltipla Tipo 2a/metabolismo , Mutação , Neoplasias/genética , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret/genética
3.
Oncogene ; 41(19): 2706-2718, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35393543

RESUMO

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.


Assuntos
Quebras de DNA de Cadeia Dupla , Inibidores de Poli(ADP-Ribose) Polimerases , Proteína BRCA1/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Recombinação Homóloga , Humanos , Fosfoproteínas/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Transposases/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
4.
Genes Cells ; 26(5): 328-335, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33624391

RESUMO

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.


Assuntos
Recombinação Homóloga/genética , Reparo de DNA por Recombinação , Sirtuína 2/metabolismo , Sirtuína 3/metabolismo , Acetilação , Quebras de DNA de Cadeia Dupla , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo
5.
Cancer Sci ; 111(5): 1443-1451, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32232911

RESUMO

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.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Instabilidade Genômica/genética , Neoplasias/genética , Transcrição Gênica , Dano ao DNA , Recombinação Homóloga , Humanos , Neoplasias/metabolismo , RNA , Ativação Transcricional
7.
PLoS Genet ; 14(3): e1007277, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29590107

RESUMO

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.


Assuntos
Quebras de DNA de Cadeia Dupla , Histona Acetiltransferases/fisiologia , Histona Desacetilases/fisiologia , Recombinação Homóloga , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Acetilação , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Histona Acetiltransferases/genética , Histona Desacetilases/genética , Humanos , Microscopia de Fluorescência , Técnicas do Sistema de Duplo-Híbrido
8.
Philos Trans R Soc Lond B Biol Sci ; 372(1731)2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28847829

RESUMO

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'.


Assuntos
Senescência Celular , Montagem e Desmontagem da Cromatina , Epigênese Genética , Regulação da Expressão Gênica , Neoplasias/genética , Nucleossomos/genética , Reparo do DNA , Humanos , Nucleossomos/metabolismo
9.
Nat Commun ; 8: 16102, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28719581

RESUMO

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.


Assuntos
Reparo do DNA , Histona Acetiltransferases/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Humanos , Fosforilação , Dímeros de Pirimidina/metabolismo , Fatores de Transcrição/metabolismo , Raios Ultravioleta
10.
Nucleus ; 7(2): 138-45, 2016 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-27310306

RESUMO

Polycomb group (PcG) repress, whereas Trithorax group (TrxG) activate transcription for tissue development and cellular proliferation, and misregulation of these factors is often associated with cancer. ENL (MLLT1) and AF9 (MLLT3) are fusion partners of Mixed Lineage Leukemia (MLL), TrxG proteins, and are factors in Super Elongation Complex (SEC). SEC controls transcriptional elongation to release RNA polymerase II, paused around transcription start site. In MLL rearranged leukemia, several components of SEC have been found as MLL-fusion partners and the control of transcriptional elongation is misregulated leading to tumorigenesis in MLL-SEC fused Leukemia. It has been suggested that unexpected collaboration of ENL/AF9-MLL and PcG are involved in tumorigenesis in leukemia. Recently, we found that the collaboration of ENL/AF9 and PcG led to a novel mechanism of transcriptional switch from elongation to repression under ATM-signaling for genome integrity. Activated ATM phosphorylates ENL/AF9 in SEC, and the phosphorylated ENL/AF9 binds BMI1 and RING1B, a heterodimeric E3-ubiquitin-ligase complex in Polycomb Repressive complex 1 (PRC1), and recruits PRC1 at transcriptional elongation sites to rapidly repress transcription. The ENL/AF9 in SEC- and PcG-mediated transcriptional repression promotes DSB repair near transcription sites. The implication of this is that the collaboration of ENL/AF9 in SEC and PcG ensures a rapid response of transcriptional switching from elongation to repression to neighboring genotoxic stresses for DSB repair. Therefore, these results suggested that the collaboration of ENL/AF9 and PcG in transcriptional control is required to maintain genome integrity and may be link to the MLL-ENL/AF9 leukemia.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Genoma/genética , Proteínas do Grupo Polycomb/metabolismo , Transcrição Gênica , Fatores de Elongação da Transcrição/metabolismo , Leucemia/genética , Leucemia/metabolismo
11.
Mol Cell ; 58(3): 468-82, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25921070

RESUMO

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.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Reparo do DNA , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia/genética , Western Blotting , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fosforilação , Complexo Repressor Polycomb 1/genética , Ligação Proteica , Interferência de RNA , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética , Transcrição Gênica
12.
Cancer Res ; 74(9): 2465-75, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24788099

RESUMO

The SWI/SNF chromatin-remodeling family contains various protein complexes, which regulate gene expression during cellular development and influence DNA damage response in an ATP- and complex-dependent manner, of which details remain elusive. Recent human genome sequencing of various cancer cells revealed frequent mutations in SWI/SNF factors, especially ARID1A, a variant subunit in the BRG1-associated factor (BAF) complex of the SWI/SNF family. We combined live-cell analysis and gene-suppression experiments to show that suppression of either ARID1A or its paralog ARID1B led to reduced nonhomologous end joining activity of DNA double-strand breaks (DSB), decreased accumulation of KU70/KU80 proteins at DSB, and sensitivity to ionizing radiation, as well as to cisplatin and UV. Thus, in contrast to transcriptional regulation, both ARID1 proteins are required for cellular resistance to various types of DNA damage, including DSB. The suppression of other SWI/SNF factors, namely SNF5, BAF60a, BAF60c, BAF155, or BAF170, exhibits a similar phenotype. Of these factors, ARID1A, ARID1B, SNF5, and BAF60c are necessary for the immediate recruitment of the ATPase subunit of the SWI/SNF complex to DSB, arguing that both ARID1 proteins facilitate the damage response of the complex. Finally, we found interdependent protein stability among the SWI/SNF factors, suggesting their direct interaction within the complex and the reason why multiple factors are frequently lost in parallel in cancer cells. Taken together, we show that cancer cells lacking in the expression of certain SWI/SNF factors, including ARID1A, are deficient in DNA repair and potentially vulnerable to DNA damage.


Assuntos
Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos , Humanos , Estabilidade Proteica , Subunidades Proteicas/metabolismo , Tolerância a Radiação , Fatores de Transcrição/metabolismo
13.
Mol Cell ; 53(4): 617-30, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24560272

RESUMO

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.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Tioléster Hidrolases/química , Proteínas de Transporte/metabolismo , Morte Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Biblioteca Gênica , Inativação Gênica , Células HeLa , Chaperonas de Histonas , Histonas/química , Recombinação Homóloga , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisina/química , Mutação , Proteínas Nucleares/metabolismo , Plasmídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Recombinação Genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina/química , Ubiquitina-Proteína Ligases
14.
Carcinogenesis ; 34(11): 2486-97, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23825154

RESUMO

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.


Assuntos
Curcumina/farmacologia , Dano ao DNA/efeitos dos fármacos , Recombinação Homóloga/efeitos dos fármacos , Neoplasias/patologia , Inibidores de Poli(ADP-Ribose) Polimerases , Transdução de Sinais/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA1/antagonistas & inibidores , Proteína BRCA1/metabolismo , Western Blotting , Pontos de Checagem do Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Radioisótopos de Cobalto , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Inibidores Enzimáticos/farmacologia , Raios gama , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Recombinação Homóloga/efeitos da radiação , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Regiões Promotoras Genéticas , Proteínas Quinases/metabolismo , Sialoglicoproteínas/antagonistas & inibidores , Sialoglicoproteínas/metabolismo , Transdução de Sinais/efeitos da radiação , Células Tumorais Cultivadas , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo
15.
Mol Cell ; 40(6): 976-87, 2010 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21172662

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Fatores de Transcrição/metabolismo , Antígenos Nucleares/metabolismo , Células Cultivadas , DNA Polimerase III/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Autoantígeno Ku , Nucleoproteínas/metabolismo , Raios Ultravioleta
16.
DNA Repair (Amst) ; 7(6): 879-89, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18396468

RESUMO

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.


Assuntos
Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Recombinação Genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sequência de Bases , DNA/efeitos dos fármacos , Primers do DNA , Metanossulfonato de Metila/farmacologia , Mutagênese Sítio-Dirigida , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteínas de Saccharomyces cerevisiae/genética
17.
Nucleic Acids Res ; 35(15): 4989-5000, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17636314

RESUMO

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.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Recombinação Genética , Proteínas de Saccharomyces cerevisiae/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona , Replicação do DNA , Endodesoxirribonucleases/fisiologia , Exodesoxirribonucleases/fisiologia , Deleção de Genes , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
EMBO Rep ; 8(7): 685-90, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17571075

RESUMO

The Saccharomyces cerevisiae RecQ-mediated genome instability (Rmi1) protein was recently identified as the third member of the slow growth suppressor 1-DNA topoisomerase III (Sgs1-Top3) complex, which is required for maintaining genomic stability. Here, we show that cells lacking RMI1 have a mitotic delay, which is partly dependent on the spindle checkpoint, and are sensitive to the microtubule depolymerizing agent benomyl. We show that rmi1 and top3 single mutants are defective in sister chromatid cohesion, and that deletion of SGS1 suppresses benomyl sensitivity and the cohesion defect in these mutant cells. Loss of RAD51 also suppresses the cohesion defect of rmi1 mutant cells. These results indicate the existence of a new pathway involving Rad51 and Sgs1-Top3-Rmi1, which leads to the establishment of sister chromatid cohesion.


Assuntos
Cromátides/metabolismo , RecQ Helicases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Cromátides/genética , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA , Citometria de Fluxo , Saccharomyces cerevisiae/genética , Troca de Cromátide Irmã
19.
Genes Genet Syst ; 82(2): 157-60, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17507781

RESUMO

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.


Assuntos
Cromossomos Fúngicos , Proteínas de Ligação a DNA/fisiologia , Metanossulfonato de Metila/farmacologia , Mitose/efeitos dos fármacos , Recombinação Genética/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/fisiologia , Homologia de Sequência , Proliferação de Células/efeitos dos fármacos , Mitose/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética
20.
Biochem Biophys Res Commun ; 356(4): 1031-7, 2007 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-17399687

RESUMO

Unequal sister chromatid recombination (uSCR) is elevated in budding yeast sgs1 mutants, which lack a homolog of the human BLM gene that causes Bloom syndrome. Examination of the mechanism responsible for elevated uSCR in sgs1 mutants showed that mutation of RAD51 also resulted in hyper-uSCR. Data from this study show that defects in the Rad51-Sgs1-dependent and Sgs1-dependent lesion-bypass pathways activate Rad59-Rad1- and Rad59-dependent pathways, respectively, resulting in uSCR. Moreover, the elevation of uSCR in sgs1 and rad51 mutants was dependent on MMS1, which encodes one of the components of the Mms22 module. Lastly, a putative role of Mms1 in the elevation of uSCR and a possible mechanism by which uSCR is elevated as a result of defective Sgs1 and Rad51 are discussed.


Assuntos
Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , RecQ Helicases/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Transdução de Sinais/fisiologia , Troca de Cromátide Irmã/fisiologia , Proteína Rad52 de Recombinação e Reparo de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...