Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 127(7): 1361-73, 2006 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-17190600

RESUMO

Histone lysine methylation has been linked to the recruitment of mammalian DNA repair factor 53BP1 and putative fission yeast homolog Crb2 to DNA double-strand breaks (DSBs), but how histone recognition is achieved has not been established. Here we demonstrate that this link occurs through direct binding of 53BP1 and Crb2 to histone H4. Using X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, we show that, despite low amino acid sequence conservation, both 53BP1 and Crb2 contain tandem tudor domains that interact with histone H4 specifically dimethylated at Lys20 (H4-K20me2). The structure of 53BP1/H4-K20me2 complex uncovers a unique five-residue 53BP1 binding cage, remarkably conserved in the structure of Crb2, that best accommodates a dimethyllysine but excludes a trimethyllysine, thus explaining the methylation state-specific recognition of H4-K20. This study reveals an evolutionarily conserved molecular mechanism of targeting DNA repair proteins to DSBs by direct recognition of H4-K20me2.


Assuntos
Proteínas de Transporte/química , Reparo do DNA , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas de Membrana/química , Fosfoproteínas/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cromatina/química , Cromatina/metabolismo , Sequência Conservada , Cristalografia por Raios X , Quebras de DNA de Cadeia Dupla , Histonas/química , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisina , Espectroscopia de Ressonância Magnética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metilação , Dados de Sequência Molecular , Mutação , Fosfoproteínas/metabolismo , Estrutura Terciária de Proteína , Sequências de Repetição em Tandem , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
2.
Mol Cell Biol ; 25(22): 10079-86, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16260621

RESUMO

p53 binding protein 1 (53BP1) is a putative DNA damage sensor that accumulates at sites of double-strand breaks (DSBs) in a manner dependent on histone H2AX. Here we show that the loss of one or both copies of 53BP1 greatly accelerates lymphomagenesis in a p53-null background, suggesting that 53BP1 and p53 cooperate in tumor suppression. A subset of 53BP1-/- p53-/- lymphomas, like those in H2AX-/- p53-/- mice, were diploid and harbored clonal translocations involving antigen receptor loci, indicating misrepair of DSBs during V(D)J recombination as one cause of oncogenic transformation. Loss of a single 53BP1 allele compromised genomic stability and DSB repair, which could explain the susceptibility of 53BP1+/- mice to tumorigenesis. In addition to structural aberrations, there were high rates of chromosomal missegregation and accumulation of aneuploid cells in 53BP1-/- p53+/+ and 53BP1-/- p53-/- tumors as well as in primary 53BP1-/- splenocytes. We conclude that 53BP1 functions as a dosage-dependent caretaker that promotes genomic stability by a mechanism that preserves chromosome structure and number.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias/genética , Fosfoproteínas/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Alelos , Motivos de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Células Cultivadas , Centrossomo/ultraestrutura , Aberrações Cromossômicas , Cromossomos/ultraestrutura , Cruzamentos Genéticos , Reparo do DNA , Feminino , Genes Supressores de Tumor , Predisposição Genética para Doença , Hibridização in Situ Fluorescente , Cariotipagem , Linfoma/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Neoplasias/patologia , Receptores de Antígenos/metabolismo , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Fatores de Tempo , Translocação Genética , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
3.
Nat Genet ; 37(4): 401-6, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793587

RESUMO

Tumorigenesis is a consequence of loss of tumor suppressors and activation of oncogenes. Expression of the mitotic checkpoint protein Chfr is lost in 20-50% of primary tumors and tumor cell lines. To explore whether downregulation of Chfr contributes directly to tumorigenesis, we generated Chfr knockout mice. Chfr-deficient mice are cancer-prone, develop spontaneous tumors and have increased skin tumor incidence after treatment with dimethylbenz(a)anthracene. Chfr deficiency leads to chromosomal instability in embryonic fibroblasts and regulates the mitotic kinase Aurora A, which is frequently upregulated in a variety of tumors. Chfr physically interacts with Aurora A and ubiquitinates Aurora A both in vitro and in vivo. Collectively, our data suggest that Chfr is a tumor suppressor and ensures chromosomal stability by controlling the expression levels of key mitotic proteins such as Aurora A.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Quinases/metabolismo , Neoplasias Cutâneas/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Aurora Quinase A , Aurora Quinases , Carcinógenos/toxicidade , Proteínas de Ciclo Celular/genética , Instabilidade Cromossômica , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Feminino , Fibroblastos/enzimologia , Marcação de Genes , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Knockout , Mitose/genética , Proteínas de Neoplasias/genética , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases , Proteínas de Xenopus
4.
J Cell Biol ; 166(6): 801-13, 2004 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-15364958

RESUMO

Bloom's syndrome is a rare autosomal recessive genetic disorder characterized by chromosomal aberrations, genetic instability, and cancer predisposition, all of which may be the result of abnormal signal transduction during DNA damage recognition. Here, we show that BLM is an intermediate responder to stalled DNA replication forks. BLM colocalized and physically interacted with the DNA damage response proteins 53BP1 and H2AX. Although BLM facilitated physical interaction between p53 and 53BP1, 53BP1 was required for efficient accumulation of both BLM and p53 at the sites of stalled replication. The accumulation of BLM/53BP1 foci and the physical interaction between them was independent of gamma-H2AX. The active Chk1 kinase was essential for both the accurate focal colocalization of 53BP1 with BLM and the consequent stabilization of BLM. Once the ATR/Chk1- and 53BP1-mediated signal from replicational stress is received, BLM functions in multiple downstream repair processes, thereby fulfilling its role as a caretaker tumor suppressor.


Assuntos
Síndrome de Bloom/enzimologia , DNA Helicases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases/metabolismo , Fase S , Western Blotting , Bromodesoxiuridina/metabolismo , Proteínas de Transporte , Linhagem Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , DNA Helicases/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Hidroxiureia/farmacologia , Cinética , Microscopia Confocal , Fosfoproteínas , Fosforilação , Testes de Precipitina , RNA Interferente Pequeno , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
5.
J Cell Biol ; 165(4): 459-64, 2004 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15159415

RESUMO

53BP1 participates early in the DNA damage response and is involved in cell cycle checkpoint control. Moreover, the phenotype of mice and cells deficient in 53BP1 suggests a defect in DNA repair (Ward et al., 2003b). Therefore, we asked whether or not 53BP1 would be required for the efficient repair of DNA double strand breaks. Our data indicate that homologous recombination by gene conversion does not depend on 53BP1. Moreover, 53BP1-deficient mice support normal V(D)J recombination, indicating that 53BP1 is not required for "classic" nonhomologous end joining. However, class switch recombination is severely impaired in the absence of 53BP1, suggesting that 53BP1 facilitates DNA end joining in a way that is not required or redundant for the efficient closing of RAG-induced strand breaks. These findings are similar to those observed in mice or cells deficient in the tumor suppressors ATM and H2AX, further suggesting that the functions of ATM, H2AX, and 53BP1 are closely linked.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fosfoproteínas , Recombinação Genética/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular , DNA/genética , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/genética , Rearranjo Gênico/genética , Histonas/deficiência , Histonas/genética , Camundongos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Homologia de Sequência do Ácido Nucleico , Proteínas Supressoras de Tumor
6.
J Biol Chem ; 279(11): 9677-80, 2004 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-14742437

RESUMO

Ataxia-telangiectasia-mutated and Rad3-related (ATR) plays an essential role in the maintenance of genome integrity and cell viability. The kinase is activated in response to DNA damage and initiates a checkpoint signaling cascade by phosphorylating a number of downstream substrates including Chk1. Unlike ataxia-telangiectasia-mutated (ATM), which appears to be mainly activated by DNA double-strand breaks, ATR can be activated by a variety of DNA damaging agents. However, it is still unclear what triggers ATR activation in response to such diverse DNA lesions. One model proposes that ATR can directly recognize DNA lesions, while other recent data suggest that ATR is activated by a common single-stranded DNA (ssDNA) intermediate generated during DNA repair. In this study, we show that UV lesions do not directly activate ATR in vivo. In addition, ssDNA lesions created during the repair of UV damage are also not sufficient to activate the ATR-dependent pathway. ATR activation is only observed in replicating cells indicating that replication stress is required to trigger the ATR-mediated checkpoint cascade in response to UV irradiation. Interestingly, H2AX appears to be required for the accumulation of ATR at stalled replication forks. Together our data suggest that ssDNA at arrested replication forks recruits ATR and initiates ATR-mediated phosphorylation of H2AX and Chk1. Phosphorylated H2AX might further facilitate ATR activation by stabilizing ATR at the sites of arrested replication forks.


Assuntos
Raios Ultravioleta , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , Reparo do DNA , Replicação do DNA , DNA de Cadeia Simples , Citometria de Fluxo , Células HeLa , Humanos , Immunoblotting , Microscopia de Fluorescência , Fosforilação , Proteínas Quinases/metabolismo , Transdução de Sinais , Estresse Fisiológico , Fatores de Tempo
7.
J Biol Chem ; 278(22): 19579-82, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12697768

RESUMO

53BP1 participates in the cellular response to DNA damage. Like many proteins involved in the DNA damage response, 53BP1 becomes hyperphosphorylated after radiation and colocalizes with phosphorylated H2AX in megabase regions surrounding the sites of DNA strand breaks. However, it is not yet clear whether the phosphorylation status of 53BP1 determines its localization or vice versa. In this study we mapped a region upstream of the 53BP1 C terminus that is required and sufficient for the recruitment of 53BP1 to these DNA break areas. In vitro assays revealed that this region binds to phosphorylated but not unphosphorylated H2AX. Moreover, using H2AX-deficient cells reconstituted with wild-type or a phosphorylation-deficient mutant of H2AX, we have shown that phosphorylation of H2AX at serine 140 is critical for efficient 53BP1 foci formation, implying that a direct interaction between 53BP1 and phosphorylated H2AX is required for the accumulation of 53BP1 at DNA break sites. On the other hand, radiation-induced phosphorylation of the 53BP1 N terminus by the ATM (ataxia-telangiectasia mutated) kinase is not essential for 53BP1 foci formation and takes place independently of 53BP1 redistribution. Thus, these two damage-induced events, hyperphosphorylation and relocalization of 53BP1, occur independently in the cell.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , DNA/metabolismo , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Fosfoproteínas , Proteínas de Transporte/genética , Linhagem Celular , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica
8.
Mol Cell Biol ; 23(7): 2556-63, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12640136

RESUMO

53BP1 is a p53 binding protein of unknown function that binds to the central DNA-binding domain of p53. It relocates to the sites of DNA strand breaks in response to DNA damage and is a putative substrate of the ataxia telangiectasia-mutated (ATM) kinase. To study the biological role of 53BP1, we disrupted the 53BP1 gene in the mouse. We show that, similar to ATM(-/-) mice, 53BP1-deficient mice were growth retarded, immune deficient, radiation sensitive, and cancer prone. 53BP1(-/-) cells show a slight S-phase checkpoint defect and prolonged G(2)/M arrest after treatment with ionizing radiation. Moreover, 53BP1(-/-) cells feature a defective DNA damage response with impaired Chk2 activation. These data indicate that 53BP1 acts downstream of ATM and upstream of Chk2 in the DNA damage response pathway and is involved in tumor suppression.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , Transtornos do Crescimento/genética , Peptídeos e Proteínas de Sinalização Intracelular , Linfoma/genética , Síndromes Neoplásicas Hereditárias/genética , Fosfoproteínas , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA , Marcação de Genes , Transtornos do Crescimento/complicações , Linfoma/complicações , Linfoma/patologia , Camundongos , Camundongos Knockout , Síndromes Neoplásicas Hereditárias/complicações , Fenótipo , Ligação Proteica/fisiologia , Tolerância a Radiação/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...