Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
DNA Cell Biol ; 26(9): 683-94, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17678437

RESUMO

Response to genotoxic stress may trigger the activation of distinct mechanisms that serve to promote cell death, including apoptosis and necrosis. In this study we examined the response of human fibroblasts, either proficient or deficient for the damage-activated protein kinase ataxia telangiectasia-mutated (ATM), to the alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Analysis of both long- and short-term viability shows that both ATM-proficient YZ-5 and ATM-deficient EBS-7 fibroblasts display a cytotoxic response to MNNG. Consistent with activation of apoptosis in response to MNNG, we observed increased caspase-3 cleavage and activity, appearance of fragmented nuclei, and increased staining with annexin V in both ATM-proficient and -deficient fibroblasts. Flow cytometry demonstrated that these cell lines also display a nonapoptotic cell death in response to MNNG. This form of cell death is associated with activation of poly-ADP ribose polymerase (PARP), and analysis of PARP activity indicated increased protein poly(ADP-ribosylation) in YZ-5 when compared to EBS-7. This PARP activity was accompanied by apoptosis-inducing factor release and translocation from the mitochondria to the nucleus. Finally, the PARP inhibitor 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ) or the caspase-3 inhibitor benzyloxycarbonyl-VAD-fluoromethyl ketone dramatically diminished the cytotoxic response to MNNG, reinforcing the roles for apoptotic and nonapoptotic cell death in human fibroblasts treated with MNNG. From these findings, we conclude that MNNG induces a heterogeneous death response in human fibroblasts.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fibroblastos/efeitos dos fármacos , Metilnitronitrosoguanidina/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Anexina A5/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , Ensaio de Unidades Formadoras de Colônias , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Humanos , Immunoblotting , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Proteínas Supressoras de Tumor/genética
2.
Mol Pharmacol ; 68(4): 1049-60, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15994368

RESUMO

S(N)1-alkylating agents, such as the mutagenic and cytotoxic drug N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), robustly activate the DNA damage-responsive G(2) checkpoint. Establishment of this checkpoint is dependent on a functional mismatch repair (MMR) system; however, exposure to high doses of MNNG overrides the requirement for MMR to trigger G(2) arrest. In addition, unlike moderate-dose exposure, in which the G(2) checkpoint is attenuated in ataxia-telangiectasia, mutated (ATM)-deficient cells, high-dose MNNG treatment activates G(2) arrest through an ATM-independent mechanism. We document that this arrest is sensitive to the pharmacological agents caffeine and 7-hydroxystaurosporine (UCN-01) that inhibit the checkpoint kinases ATM/ATM and Rad-3-related (ATR) and Chk1/Chk2, respectively. Furthermore, these agents block inactivation of the cell-cycle regulatory molecules Cdc25C and Cdc2, establishing the downstream mechanism through which high-dose MNNG establishes G(2) arrest. Activation of both Chk2 and Chk1 was independent of ATM and MMR in response to high-dose MNNG, unlike the response to moderate doses of this drug. Chk2 was found to be dispensable for cell-cycle arrest in response to high-dose MNNG treatment; however, ATR deficiency and decreased Chk1 expression forced by RNA interference resulted in diminished checkpoint response. These results indicate that MNNG activates the G(2) checkpoint through different mechanisms activated in a dose-dependent fashion.


Assuntos
Ciclo Celular/efeitos dos fármacos , Metilnitronitrosoguanidina/farmacologia , Linhagem Celular Transformada , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Relação Dose-Resposta a Droga , Humanos , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
3.
Mol Biol Cell ; 16(3): 1513-26, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15647386

RESUMO

SN1 DNA methylating agents such as the nitrosourea N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) elicit a G2/M checkpoint response via a mismatch repair (MMR) system-dependent mechanism; however, the exact nature of the mechanism governing MNNG-induced G2/M arrest and how MMR mechanistically participates in this process are unknown. Here, we show that MNNG exposure results in activation of the cell cycle checkpoint kinases ATM, Chk1, and Chk2, each of which has been implicated in the triggering of the G2/M checkpoint response. We document that MNNG induces a robust, dose-dependent G2 arrest in MMR and ATM-proficient cells, whereas this response is abrogated in MMR-deficient cells and attenuated in ATM-deficient cells treated with moderate doses of MNNG. Pharmacological and RNA interference approaches indicated that Chk1 and Chk2 are both required components for normal MNNG-induced G2 arrest. MNNG-induced nuclear exclusion of the cell cycle regulatory phosphatase Cdc25C occurred in an MMR-dependent manner and was compromised in cells lacking ATM. Finally, both Chk1 and Chk2 interact with the MMR protein MSH2, and this interaction is enhanced after MNNG exposure, supporting the notion that the MMR system functions as a molecular scaffold at the sites of DNA damage that facilitates activation of these kinases.


Assuntos
Pareamento Incorreto de Bases , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Fase G2 , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Estaurosporina/análogos & derivados , Divisão Celular , Núcleo Celular/metabolismo , Células Cultivadas , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Dano ao DNA , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Metilnitronitrosoguanidina/farmacologia , Proteína 2 Homóloga a MutS , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estaurosporina/farmacologia , Frações Subcelulares , Fatores de Tempo
4.
Toxicol Appl Pharmacol ; 202(1): 84-98, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15589979

RESUMO

One of the cellular responses to DNA damaging events is the activation of programmed cell death, also known as apoptosis. Apoptosis is an important process in limiting tumorigenesis by eliminating cells with damaged DNA. This view is reinforced by the finding that many genes with pro-apoptotic function are absent or altered in cancer cells. The tumor suppressor p53 performs a significant role in apoptotic signaling by controlling expression of a host of genes that have pro-apoptotic or pro-survival function. The S(N)1 DNA alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) triggers apoptosis and the upregulation/phosphorylation of p53; however, the mechanism(s) governing MNNG-induced cell death remain unresolved. We observed that the human lymphoblastoid cell line WTK-1, which expresses mutant p53, shows far less sensitivity to the cytotoxic effects of MNNG than the closely related, p53-normal line TK-6. Exposure to 15 muM MNNG (LD50 at 24 h in TK-6) leads to a kinetically slower rate of apoptotic onset in WTK-1 cells compared to TK-6 as judged by viability assays and approaches that directly examine apoptotic onset. Similar results were obtained using an unrelated human lymphoblastoid line B310 expressing reduced levels of p53 due to E6 oncoprotein expression, indicating that MNNG activates both p53-dependent and -independent apoptotic mechanisms and that these two mechanisms are discernable by the rates which they trigger apoptotic onset. We document, during time points corresponding to peak apoptotic response in TK6, WTK-1, B310, and B310-E6, that these cell lines show marked decreases in mitochondrial transmembrane potential and increases in cytochrome c within the cytosolic fraction of MNNG-treated cells. Consistent with these events, we observed that both caspase-9 and -3 are activated in our panel of lymphoblastoid cells after MNNG exposure. We also found, using both broad spectrum and specific inhibitors, that blocking caspase activity in TK-6 and B310 cells had a significant effect on apoptotic advance, but that this treatment had no effect on entry of WTK-1 or B310-E6 cells into apoptosis. Finally, the PARP inhibitors benzamide and 6(5H)-phenanthridinone exerted notable inhibition of PARP activity and the nuclear translocation of the mitochondrial protein AIF (apoptosis-inducing factor) in MNNG-treated cells; however, these compounds exhibited no detectable inhibitory effects on MNNG-induced death in human lymphoblastoid cells. These observations suggest that PARP activity is not required during MNNG-triggered apoptosis in this cell type. Taken together, our observations support the conclusion that MNNG activates multiple apoptogenic pathways that contain both common and unique mechanisms.


Assuntos
Apoptose/efeitos dos fármacos , Metilnitronitrosoguanidina/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Benzamidas/farmacologia , Caspase 3 , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Quinase do Ponto de Checagem 2 , Humanos , Mitocôndrias/efeitos dos fármacos , Fenantrenos/farmacologia , Poli(ADP-Ribose) Polimerases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia
5.
Exp Cell Res ; 290(2): 402-13, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14567997

RESUMO

Positional cloning of the cancer-associated 20q13.2 amplicon identified two genes that display high mRNA levels in breast tumors and here we report the initial characterization of one of these gene products, designated Novel Amplified in Breast Cancer-1 (NABC1). Analysis of the primary structure of the NABC1 protein uncovered two regions of this protein with a high likelihood of forming coiled-coils and assembly of a mouse NABC1 cDNA showed that this protein is conserved between mouse and man. NABC1 antisera showed that, like its transcript, breast tumor lines that harbor amplification of 20q13.2 display high levels of the NABC1 protein compared to normal human fibroblasts or a breast cancer line that does not overexpress the NABC1 mRNA. Further, we conclude from studies using in vivo and in vitro approaches that the NABC1 protein forms detergent stable homodimers, and it is this homodimeric form that accumulates in cells that overexpress this protein. NABC1 mRNA exhibits a limited expression pattern in human tissue with high relative transcript levels observed only in brain and prostate. Immunofluorescence microscopy indicates NABC1 displays a punctate localization pattern in the cytoplasm of cultured cells, but biochemical fractionation indicates that this protein is not an integral component of membranous cytoplasmic organelles. Finally, overexpression of human NABC1 in mouse NIH/3T3 cells did not affect either the growth rate or anchorage-dependent growth properties, suggesting that NABC1 is not a prototypical oncogene.


Assuntos
Fibroblastos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Animais , Baculoviridae , Northern Blotting , Western Blotting , Linhagem Celular Transformada , Transformação Celular Viral , Citoplasma/metabolismo , Imunofluorescência , Amplificação de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Células NIH 3T3/metabolismo , RNA Mensageiro/metabolismo , Coelhos , Vírus 40 dos Símios , Transcrição Gênica
6.
Nat Genet ; 33(1): 80-4, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12447371

RESUMO

Defective S-phase checkpoint activation results in an inability to downregulate DNA replication following genotoxic insult such as exposure to ionizing radiation. This 'radioresistant DNA synthesis' (RDS) is a phenotypic hallmark of ataxia-telangiectasia, a cancer-prone disorder caused by mutations in ATM. The mismatch repair system principally corrects nucleotide mismatches that arise during replication. Here we show that the mismatch repair system is required for activation of the S-phase checkpoint in response to ionizing radiation. Cells deficient in mismatch repair proteins showed RDS, and restoration of mismatch repair function restored normal S-phase checkpoint function. Catalytic activation of ATM and ATM-mediated phosphorylation of the protein NBS1 (also called nibrin) occurred independently of mismatch repair. However, ATM-dependent phosphorylation and activation of the checkpoint kinase CHK2 and subsequent degradation of its downstream target, CDC25A, was abrogated in cells lacking mismatch repair. In vitro and in vivo approaches both show that MSH2 binds to CHK2 and that MLH1 associates with ATM. These findings indicate that the mismatch repair complex formed at the sites of DNA damage facilitates the phosphorylation of CHK2 by ATM, and that defects in this mechanism form the molecular basis for the RDS observed in cells deficient in mismatch repair.


Assuntos
Pareamento Incorreto de Bases/genética , Reparo do DNA , Proteínas de Ligação a DNA , Proteínas Proto-Oncogênicas , Tolerância a Radiação/genética , Fase S , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte , Proteínas de Ciclo Celular/metabolismo , Quinase do Ponto de Checagem 2 , Reparo do DNA/efeitos da radiação , Replicação do DNA/efeitos da radiação , Ativação Enzimática , Deleção de Genes , Humanos , Modelos Biológicos , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Radiação Ionizante , Fase S/efeitos da radiação , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Fosfatases cdc25/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA