Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Nat Genet ; 33(1): 80-4, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12447371

RESUMEN

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.


Asunto(s)
Disparidad de Par Base/genética , Reparación del ADN , Proteínas de Unión al ADN , Proteínas Proto-Oncogénicas , Tolerancia a Radiación/genética , Fase S , Proteínas Adaptadoras Transductoras de Señales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas Portadoras , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2 , Reparación del ADN/efectos de la radiación , Replicación del ADN/efectos de la radiación , Activación Enzimática , Eliminación de Gen , Humanos , Modelos Biológicos , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Radiación Ionizante , Fase S/efectos de la radiación , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Fosfatasas cdc25/metabolismo
2.
Mol Biol Cell ; 16(3): 1513-26, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15647386

RESUMEN

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.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Fase G2 , Proteínas Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Estaurosporina/análogos & derivados , División Celular , Núcleo Celular/metabolismo , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Daño del ADN , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Immunoblotting , Inmunoprecipitación , Metilnitronitrosoguanidina/farmacología , Proteína 2 Homóloga a MutS , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Estaurosporina/farmacología , Fracciones Subcelulares , Factores de Tiempo
3.
DNA Cell Biol ; 26(9): 683-94, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17678437

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Fibroblastos/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/fisiología , Anexina A5/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Caspasas/metabolismo , Proteínas de Ciclo Celular/genética , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , Ensayo de Unidades Formadoras de Colonias , Proteínas de Unión al ADN/genética , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Humanos , Immunoblotting , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Proteínas Supresoras de Tumor/genética
4.
Mol Pharmacol ; 68(4): 1049-60, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15994368

RESUMEN

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.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Línea Celular Transformada , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
5.
Toxicol Appl Pharmacol ; 202(1): 84-98, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15589979

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Proteína p53 Supresora de Tumor/fisiología , Benzamidas/farmacología , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Línea Celular , Quinasa de Punto de Control 2 , Humanos , Mitocondrias/efectos de los fármacos , Fenantrenos/farmacología , Poli(ADP-Ribosa) Polimerasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología
6.
Exp Cell Res ; 290(2): 402-13, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14567997

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Animales , Baculoviridae , Northern Blotting , Western Blotting , Línea Celular Transformada , Transformación Celular Viral , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente , Amplificación de Genes , Regulación de la Expresión Génica , Humanos , Ratones , Células 3T3 NIH/metabolismo , ARN Mensajero/metabolismo , Conejos , Virus 40 de los Simios , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA