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










Base de datos
Intervalo de año de publicación
1.
J Natl Cancer Inst ; 93(20): 1534-40, 2001 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-11604476

RESUMEN

BACKGROUND: Defective mismatch repair (MMR) in humans is particularly associated with familial colorectal cancer, but defective repair in mice is generally associated with lymphoma in the absence of experimental exposure to carcinogens. Loss of MMR also confers resistance to the toxic effects of methylating agents. We investigated whether resistance to methylation contributes to increased susceptibility to colorectal cancer in mice by exposing mice with defects in the MMR gene msh2 to a methylating agent. METHODS: Tumor incidence and time of death in msh2(+/+), msh2(+/-), and msh2(-/-) mice were analyzed after weekly exposure (until tumor appearance) to the methylating agent 1,2-dimethylhydrazine (DMH). Chemically induced and spontaneous tumors were characterized by frequency, type, and location. The tumor incidence in untreated and treated mice of each genotype was compared by a Mann-Whitney U test. Carcinogen-induced apoptosis in histologic sections of small and large intestines was also determined. All statistical tests were two-sided. RESULTS: Homozygous inactivation of the msh2 gene statistically significantly accelerated (P<.0001) death due to the development of DMH-induced colorectal tumors and lymphomas. Rates of death from DMH-induced colorectal adenocarcinoma were similar in msh2 heterozygous and wild-type mice, but only msh2 heterozygotes (msh(+/-)) developed additional, noncolorectal malignancies (notably trichofolliculoma [two of 21], angiosarcoma of the kidney capsule [two of 21], and lymphoma [one of 21]), suggesting that heterozygosity for msh2 slightly increases DMH susceptibility. DMH induced apoptosis in small intestinal and colonic epithelial crypts that was dependent on active msh2. CONCLUSIONS: Inactivation of msh2 allows the proliferation of gastrointestinal tract cells damaged by methylating agents. Furthermore, MMR constitutes a powerful defense against colorectal cancer induced by DNA methylation.


Asunto(s)
1,2-Dimetilhidrazina/toxicidad , Adenocarcinoma/genética , Alquilantes/toxicidad , Carcinógenos/toxicidad , Neoplasias del Colon/genética , Reparación del ADN/genética , Proteínas de Unión al ADN , Linfoma no Hodgkin/genética , Proteínas Proto-Oncogénicas/fisiología , Adenocarcinoma/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Disparidad de Par Base , División Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/inducido químicamente , Resistencia a Medicamentos , Genotipo , Enfermedades del Cabello/inducido químicamente , Enfermedades del Cabello/genética , Folículo Piloso , Hemangiosarcoma/inducido químicamente , Hemangiosarcoma/genética , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/patología , Neoplasias Renales/inducido químicamente , Neoplasias Renales/genética , Linfoma no Hodgkin/inducido químicamente , Metilación , Ratones , Ratones Noqueados , Proteína 2 Homóloga a MutS , Neoplasias Basocelulares/inducido químicamente , Neoplasias Basocelulares/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética
2.
J Clin Oncol ; 19(4): 1008-14, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11181663

RESUMEN

PURPOSE: The aim of this study was to define the prognostic role of microsatellite status in 65 stage I-II primary sporadic endometrioid endometrial adenocarcinoma (EEA) patients. PATIENTS AND METHODS: Familiarity for neoplasia was ascertained in all patients on the basis of a questionnaire. Microsatellite status was assessed by matching normal and tumoral DNA probed for five dinucleotide repeats and one mononucleotide repeat marker. Microsatellite status was analyzed in relation to clinicopathologic characteristics of the patients and length of disease-free survival (DFS). RESULTS: Eleven tumors (17%) of 65 had instability at two or more loci and were considered as unstable or microsatellite instability (MI). Tumors with no instability or instability at one locus were classified as microsatellite stable (MS). The percentage of MI was significantly higher in poorly than in well to moderately differentiated tumors (50% v 9%; P =.003). The 5-year DFS rate of MI patients was 63% (95% confidence interval [CI], 35% to 91%) versus 96% (95% CI, 91% to 101%) of MS patients (P =.0004). In multivariate analysis, only the presence of MI, stage II of disease, and depth of myometrial invasion greater than 50% retained independent prognostic roles. CONCLUSION: The assessment of microsatellite status may provide useful information for preoperative prognostic characterization of stage I-II primary sporadic EEA patients in which more individualized treatment options can be attempted.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Endometriales/genética , Repeticiones de Microsatélite/genética , Proteínas Adaptadoras Transductoras de Señales , Factores de Edad , Proteínas Portadoras , Femenino , Humanos , Persona de Mediana Edad , Análisis Multivariante , Homólogo 1 de la Proteína MutL , Proteínas de Neoplasias/análisis , Recurrencia Local de Neoplasia , Proteínas Nucleares , Pronóstico
3.
EMBO J ; 19(7): 1711-8, 2000 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-10747038

RESUMEN

A previously unrecognized mismatch repair activity is described. Extracts of immortalized MSH2-deficient mouse fibroblasts did not correct most single base mispairs. The same extracts carried out efficient repair of A/C mismatches. A/G mispairs were less efficiently corrected and there was no significant repair of A/A. MLH1-defective mouse extracts also repaired an A/C mispair. A/C correction by Msh2(-/-) mouse cell extracts was not affected by antibodies against the PMS2 protein, which inhibited long-patch mismatch repair. A/C repair activity is thus independent of MutSalpha, MutSbeta and MutLalpha. A/C mismatches were corrected 5-fold more efficiently by extracts of Msh2 knockout mouse cells than by comparable extracts prepared from hMSH2- or hMLH1-deficient human cells. MSH2-independent A/C correction by mouse cell extracts did not require a nick in the circular duplex DNA substrate. Repair involved replacement of the A and was associated with the resynthesis of a limited stretch of

Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas de Unión al ADN , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Proteínas Portadoras , Línea Celular , Cartilla de ADN/genética , Células HeLa , Humanos , Ratones , Ratones Noqueados , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
4.
Int J Cancer ; 85(4): 590-6, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10699935

RESUMEN

Together with tolerance to killing induced by methylating agents, loss of mismatch repair (MMR) has previously been found to be associated with hypersensitivity to the DNAcross-linking agent 1-(2-chloroethyl)-3-cyclohexyl-nitrosourea(CCNU) in several human tumor cell lines (Aquilina et al., 1998). Here, we have investigated whether MMR might act as an efficient repair pathway and provide protection against the clastogenicity induced by CCNU and whether the hypersensitivity of MMR-defective cells is extended to other cross-linking agents. An increase in cell killing and in the frequency of micronuclei was observed after CCNU exposure in 2 hPMS2-defective clones (clones 6 and 7) compared with the parental HeLa cells. Introduction of a wild-type copy of chromosome 7 in clone 7 led to re-expression of the hPMS2 protein and brought survival and chromosomal damage upon CCNU exposure to parental levels. Our data indicate that MMR protects against the clastogenic damage induced by this drug. The hPMS2-defective HeLa cells were also hypersensitive to killing by mitomycin C. Mitomycin C sensitivity was confirmed in an hMLH1-defective clone derived from Raji cells and in msh2-defective mouse embryo fibroblasts derived from knock-out mice. hPMS2-defective and parental HeLa cells were transplanted into nude mice, and the animals were treated with mitomycin C. While parental cell growth rate was unaffected, the growth of MMR-defective tumor was significantly reduced. Our results indicate that the in vitro hypersensitivity to mitomycin C conferred by loss of MMR is paralleled in vivo and may have implications for the chemotherapy of MMR-defective tumors.


Asunto(s)
Adenosina Trifosfatasas , Disparidad de Par Base , Reactivos de Enlaces Cruzados/toxicidad , Enzimas Reparadoras del ADN , Proteínas de Unión al ADN , Lomustina/toxicidad , Mitomicina/toxicidad , Proteínas/genética , Animales , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/patología , División Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/uso terapéutico , Células HeLa , Humanos , Lomustina/uso terapéutico , Melfalán/uso terapéutico , Melfalán/toxicidad , Ratones , Ratones Noqueados , Ratones Desnudos , Pruebas de Micronúcleos , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Mitomicina/uso terapéutico , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
5.
Carcinogenesis ; 20(2): 205-14, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10069455

RESUMEN

The long-patch mismatch repair pathway contributes to the cytotoxic effect of methylating agents and loss of this pathway confers tolerance to DNA methylation damage. Two methylation-tolerant mouse cell lines were identified and were shown to be defective in the MSH2 protein by in vitro mismatch repair assay. A normal copy of the human MSH2 gene, introduced by transfer of human chromosome 2, reversed the methylation tolerance. These mismatch repair defective mouse cells together with a fibroblast cell line derived from an MSH2-/- mouse, were all as resistant to N-methyl-N-nitrosourea as repair-defective human cells. Although long-patch mismatch repair-defective human cells were 50- to 100-fold more resistant to methylating agents than repair-proficient cells, loss of the same pathway from mouse cells conferred only a 3-fold increase. This discrepancy was accounted for by the intrinsic N-methyl-N-nitrosourea resistance of normal or transformed mouse cells compared with human cells. The >20-fold differential resistance between mouse and human cells could not be explained by the levels of either DNA methylation damage or the repair enzyme O6-methylguanine-DNA methyltransferase. The resistance of mouse cells to N-methyl-N-nitrosourea was selective and no cross-resistance to unrelated DNA damaging agents was observed. Pathways of apoptosis were apparently intact and functional after exposure to either N-methyl-N-nitrosourea or ultraviolet light. Extracts of mouse cells were found to perform 2-fold less long-patch mismatch repair. The reduced level of mismatch repair may contribute to their lack of sensitivity to DNA methylation damage.


Asunto(s)
Alquilantes/farmacología , Disparidad de Par Base/genética , Metilación de ADN , Reparación del ADN , Proteínas de Unión al ADN , Metilnitrosourea/farmacología , Células 3T3/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis , Disparidad de Par Base/efectos de los fármacos , Cromosomas Humanos Par 2/genética , Resistencia a Antineoplásicos/genética , Técnicas de Transferencia de Gen , Genotipo , Guanina/análogos & derivados , Guanina/farmacología , Células HeLa/efectos de los fármacos , Humanos , Melanoma Experimental , Metilnitronitrosoguanidina , Ratones , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogénicas/genética , Especificidad de la Especie , Células Tumorales Cultivadas/efectos de los fármacos
6.
Mutat Res ; 385(2): 115-26, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9447233

RESUMEN

Human cell lines resistant to N-methyl-N-nitrosourea (MNU) were previously assigned to two complementation groups. Members of group I are defective in mismatch correction [S. Ceccotti, G Aquilina, P. Macpherson, M. Yamada, P. Karran, M. Bignami, Processing of O6-methylguanine by mismatch correction in human cell extracts. Current Biol. 6 (1996) 1528-1531]. To identify the mechanism responsible for the less pronounced phenotype of the second complementation group, we characterized the persistence of MNU-induced O6-methylguanine (O6-meGua) and mutation induction at the hypoxanthine guanine phosphoribosyl-transferase (HPRT) locus. Group II clones are unable to repair the premutagenic base O6-meGua and are as mutable by MNU as group I clones and the parental HeLaMR cells. MNU-induced SCE were undetectable in group I clones and drastically reduced in group II in comparison with the parental cells. These observations are consistent with a defective processing of DNA methylation damage by members of both groups. Group II clones exhibit a moderate spontaneous mutator phenotype at the HPRT gene but significant instability at mononucleotide repeat microsatellites. Introduction of a single human chromosome 2 (but not of chromosome 3 or 7) into group II cells partially reverts both MNU resistance and the increased spontaneous mutation rate. The properties of group II variants are consistent with methylation tolerance and a partially defective mismatch repair. We propose that members of group II are defective in the chromosome 2-based mismatch correction gene GTBP/hMSH6.


Asunto(s)
Cromosomas Humanos Par 2/genética , Metilación de ADN , Reparación del ADN/genética , Centrómero , Sondas de ADN , Resistencia a Medicamentos , Técnicas de Transferencia de Gen , Guanina/análogos & derivados , Guanina/análisis , Células HeLa , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Metilnitrosourea/farmacología , Repeticiones de Microsatélite/genética , Mutagénesis , Mutágenos/farmacología , Intercambio de Cromátides Hermanas
7.
Cancer Res ; 55(12): 2569-75, 1995 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-7780970

RESUMEN

Sixty % of clones isolated from HeLa cells treated with toxic concentrations of a methylating carcinogen showed increased resistance to the cytotoxicity of N-methyl-N-nitrosourea. D37 values were 6- to 100-fold higher than in the parental cell population. The absence of detectable levels of the repair enzyme O6-methylguanine-DNA methyltransferase indicated that the resistant clones were able to tolerate the presence of O6-methylguanine in their DNA. Analysis of N-methyl-N-nitrosourea survival in the hybrids between tolerant clones and HeLa cells showed that tolerance can be either recessive or codominant. Fusion between tolerant clones indicated two complementation groups. We measured spontaneous mutation rates at microsatellites and at the hypoxanthine-guanine phosphoribosyl transferase (hprt) locus in several tolerant clones. All the clones of Complementation Group I showed unstable microsatellites and 4-8-fold increases in mutation rates at hprt. No significant alterations in spontaneous mutation rates were found in clones of Complementation Group II. The data indicate that tolerance to methylation damage can be conferred by alterations in at least two different gene products and that one of the two groups has the mutator phenotype typical of mismatch correction defective cells.


Asunto(s)
Daño del ADN , Prueba de Complementación Genética , Metilnitrosourea/toxicidad , Mutagénesis , Tioguanina/toxicidad , Supervivencia Celular/efectos de los fármacos , Reparación del ADN , ADN de Neoplasias/análisis , ADN de Neoplasias/química , ADN Satélite/genética , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos/genética , Guanina/análogos & derivados , Guanina/análisis , Células HeLa , Humanos , Metilación , Metiltransferasas/análisis , O(6)-Metilguanina-ADN Metiltransferasa , Fenotipo
8.
Biochem Biophys Res Commun ; 205(2): 1236-42, 1994 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-7802655

RESUMEN

The cDNA coding for the Xenopus laevis homolog of the transcriptional activator/repressor protein delta/YY1 was isolated from a lambda gt11 oocyte cDNA library. The deduced aminoacid sequence shows that the four zinc fingers of the DNA binding domain are 99% conserved when compared to the mouse (delta) and 95% to the human (YY1) proteins, while differences are found in the N-terminal region. In particular, the long run of consecutive glycines and histidines of delta and YY1 is missing. The protein, named FIII/YY1, was overexpressed into Xenopus oocytes from the cDNA under direction of the L14 rp-promoter and found to share antigenic and DNA-binding properties with the oocyte endogenous protein binding to the first exon of the X.laevis ribosomal protein genes (rp-genes) L1 and L14.


Asunto(s)
Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Exones , Proteínas Represoras/metabolismo , Proteínas Ribosómicas/genética , Factores de Transcripción/metabolismo , Xenopus laevis/genética , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Unión Competitiva , Cloranfenicol O-Acetiltransferasa/biosíntesis , Clonación Molecular , Proteínas de Unión al ADN/biosíntesis , Factores de Unión al ADN Específico de las Células Eritroides , Femenino , Expresión Génica , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Oocitos/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Mapeo Restrictivo , Homología de Secuencia de Aminoácido , Factores de Transcripción/biosíntesis , Proteínas de Xenopus , Factor de Transcripción YY1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...