Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 98(21): 12044-9, 2001 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-11593014

RESUMO

Chromatid catenation is actively monitored in human cells, with progression from G(2) to mitosis being inhibited when chromatids are insufficiently decatenated. Mitotic delay was quantified in normal and checkpoint-deficient human cells during treatment with ICRF-193, a topoisomerase II catalytic inhibitor that prevents chromatid decatenation without producing topoisomerase-associated DNA strand breaks. Ataxia telangiectasia (A-T) cells, defective in DNA damage checkpoints, showed normal mitotic delay when treated with ICRF-193. The mitotic delay in response to ICRF-193 was ablated in human fibroblasts expressing an ataxia telangiectasia mutated- and rad3-related (ATR) kinase-inactive ATR allele (ATR(ki)). BRCA1-mutant HCC1937 cells also displayed a defect in ICRF-193-induced mitotic delay, which was corrected by expression of wild-type BRCA1. Phosphorylations of hCds1 or Chk1 and inhibition of Cdk1 kinase activity, which are elements of checkpoints associated with DNA damage or replication, did not occur during ICRF-193-induced mitotic delay. Over-expression of cyclin B1 containing a dominant nuclear localization signal, and inhibition of Crm1-mediated nuclear export, reversed ICRF-193-induced mitotic delay. In combination, these results imply that ATR and BRCA1 enforce the decatenation G(2) checkpoint, which may act to exclude cyclin B1/Cdk1 complexes from the nucleus. Moreover, induction of ATR(ki) produced a 10-fold increase in chromosomal aberrations, further emphasizing the vital role for ATR in genetic stability.


Assuntos
Proteína BRCA1/metabolismo , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular , Ciclina B/metabolismo , Mitose/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Inibidores da Topoisomerase II , Ataxia Telangiectasia , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular , Núcleo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Ciclina B1 , Proteínas de Ligação a DNA , Dicetopiperazinas , Fase G2 , Humanos , Mitose/efeitos dos fármacos , Fosforilação , Piperazinas/farmacologia , Proteínas Quinases/metabolismo , Proteínas Supressoras de Tumor
2.
Cancer Res ; 58(9): 1993-2002, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9581844

RESUMO

The cyclin-dependent kinase inhibitor p21(WAF1/CIP1/SDI1/CAP20) exists in normal human fibroblasts in a quaternary complex with a cyclin, a cyclin-dependent kinase, and proliferating cell nuclear antigen. A model was proposed in which, during p53-mediated suppression of cell proliferation following treatment with 254 nm UV radiation (UVC), the enhanced expression of p21 might inhibit DNA replication by virtue of its interactions with proliferating cell nuclear antigen. To test this model, we examined the mechanisms of inhibition of DNA replication in diploid human fibroblasts that express human papillomavirus type 16 E6, which inactivates p53. E6-expressing cells were defective in G1 checkpoint responses of induction of p21 and G1 arrest after ionizing radiation-induced damage to DNA. Accordingly, E6-expressing cells were resistant to inactivation of single-cell colony formation by ionizing radiation. E6 cells also displayed normal S-phase checkpoint responses of inhibition and recovery of replicon initiation following exposure to ionizing radiation and normal ability to bypass pyrimidine dimers during DNA replication soon after UVC irradiation (i.e., postreplication repair). However, DNA replication 6 h after UVC exposure was significantly inhibited in E6 cells in comparison to isogenic controls. This failure to maintain DNA replication in S-phase cells was associated with enhanced sensitivity to inactivation of single-cell colony formation by UVC. These results indicate that the p53-induced p21 pathway is not involved in the immediate S-phase responses to radiation-induced DNA damage of inhibition of replicon initiation and translesion bypass. However, our results demonstrate that p53 and, conceivably, p21 contribute to the ability of normal human fibroblasts to sustain DNA replication activity and form colonies following UVC irradiation.


Assuntos
Replicação do DNA , Fibroblastos/fisiologia , Proteínas Repressoras , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Linhagem Celular , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Dano ao DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Inibidores Enzimáticos/metabolismo , Fibroblastos/efeitos da radiação , Fibroblastos/virologia , Citometria de Fluxo , Humanos , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/fisiologia , Fase S/efeitos da radiação , Raios Ultravioleta
3.
Toxicol Appl Pharmacol ; 141(1): 264-71, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8917699

RESUMO

The p53 gene regulates the G1 cell cycle checkpoint in response to DNA damage. A primary murine mesothelial cell line (D9) spontaneously acquired a point mutation at codon 135 in exon 5 of the p53 gene, resulting in substitution of alanine for proline; early passage D9 cells expressed wild-type p53. The growth rate of late passage D9 cells that acquired the p53 mutation was increased compared to that of early passage cells; however, this mutation was not sufficient to confer tumorigenicity to this cell line. Mammalian cells that express wild-type p53 show a transient arrest in G1 after exposure to ionizing radiation. Early passage D9 cells showed a G1 arrest following ionizing radiation, while late passage D9 cells arrested in G2 or mitosis. The clastogenic effects of ionizing radiation can be demonstrated by the cytokinesis-arrested micronucleus assay. Following treatment with cytochalasin B to arrest cytokinesis, ionizing radiation induced micronuclei in 50% of late passage D9 cells compared to 15% of early passage cells. After exposure to 15 micrograms/cm2 of crocidolite asbestos fibers, 18% of late passage cells had micronuclei compared to 4% of early passage cells. It is hypothesized that loss of the G1 cell cycle checkpoint contributes to genetic instability in murine mesothelial cells.


Assuntos
Amianto/toxicidade , Carcinógenos/toxicidade , Dano ao DNA/genética , Genes p53/efeitos dos fármacos , Genes p53/efeitos da radiação , Mutação Puntual , Radiação Ionizante , Animais , Ciclo Celular/efeitos da radiação , Linhagem Celular , Epitélio/efeitos dos fármacos , Epitélio/efeitos da radiação , Camundongos , Testes para Micronúcleos , Testes de Mutagenicidade , Análise de Sequência de DNA
4.
Environ Health Perspect ; 102 Suppl 10: 131-6, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7705288

RESUMO

Asbestos fibers have been shown to generate reactive oxygen species using a variety of in vitro assays. It is hypothesized that these highly reactive metabolites mediate the development of malignant mesothelioma induced by asbestos fibers. DNA is a potential target of oxidant attack. Adaptive responses to oxidant injury have been described during exposure of mesothelial cells to asbestos fibers in vitro. Failure of these adaptive responses may lead to genetic instability and alterations in oncogenes and tumor suppressor genes that confer a proliferative advantage to emerging neoplastic mesothelial cells.


Assuntos
Amianto , Mesotelioma/etiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Amianto/farmacologia , Dano ao DNA , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...