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










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 32(8): 968-75, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22484423

RESUMEN

Inhibition of homologous recombination (HR) is believed to be a transactivation-independent function of p53 that protects from genetic instability. Misrepair by HR can lead to genetic alterations such as translocations, duplications, insertions and loss of heterozygosity, which all bear the risk of driving oncogenic transformation. Regulation of HR by wild-type p53 (wtp53) should prevent these genomic rearrangements. Mutation of p53 is a frequent event during carcinogenesis. In particular, dominant-negative mutants inhibiting wtp53 expressed from the unperturbed allel can drive oncogenic transformation by disrupting the p53-dependent anticancer barrier. Here, we asked whether the hot spot mutants R175H and R273H relax HR control in p53-proficient cells. Utilizing an I-SceI-based reporter assay, we observed a moderate (1.5 × ) stimulation of HR upon expression of the mutant proteins in p53-proficient CV-1, but not in p53-deficient H1299 cells. Importantly, the stimulatory effect was exactly paralleled by an increase in the number of HR competent S- and G2-phase cells, which can well explain the enhanced recombination frequencies. Furthermore, the impact on HR exerted by the transactivation domain double-mutant L22Q/W23S and mutant R273P, both of which were reported to regulate HR independently of G1-arrest execution, is also exactly mirrored by cell-cycle behavior. These results are in contrast to previous concepts stating that the transactivation-independent impact of p53 on HR is a general phenomenon valid for replication-associated and also for directly induced double-strand break. Our data strongly suggest that the latter is largely mediated by cell-cycle regulation, a classical transactivation-dependent function of p53.


Asunto(s)
Roturas del ADN de Doble Cadena , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Proteína p53 Supresora de Tumor/genética , Animales , Puntos de Control del Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Fase G2/genética , Recombinación Homóloga , Humanos , Fase S/genética , Transfección , Proteína p53 Supresora de Tumor/metabolismo
2.
J Biol Chem ; 276(41): 38076-83, 2001 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-11502743

RESUMEN

DNA polymerase alpha-primase (pol-prim) is the only enzyme that can start DNA replication de novo. The 180-kDa (p180) and 68-kDa (p68) subunits of the human four-subunit enzyme are phosphorylated by Cyclin-dependent kinases (Cdks) in a cell cycle-dependent manner. Cyclin A-Cdk2 physically interacts with pol-prim and phosphorylates N-terminal amino acids of the p180 and the p68 subunits, leading to an inhibition of pol-prim in initiating cell-free SV40 DNA replication. Mutation of conserved putative Cdk phosphorylation sites in the N terminus of human p180 and p68 reduced their phosphorylation by Cyclin A-Cdk2 in vitro. In contrast to wild-type pol-prim these mutants were no longer inhibited by Cyclin A-Cdk2 in the initiation of viral DNA replication. Importantly, rather than inhibiting it, Cyclin A-Cdk2 stimulated the initiation activity of pol-prim containing a triple N-terminal alanine mutant of the p180 subunit. Together these results suggest that Cyclin A-Cdk2 executes both stimulatory and inhibitory effects on the activity of pol-prim in initiating DNA replication.


Asunto(s)
Quinasas CDC2-CDC28 , ADN Polimerasa I/metabolismo , ADN Primasa/metabolismo , Replicación del ADN , Animales , Células Cultivadas , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , ADN Polimerasa I/química , ADN Primasa/química , Humanos , Insectos , Fosforilación , Pruebas de Precipitina , Proteínas Serina-Treonina Quinasas/metabolismo
3.
Mol Cell Biol ; 21(7): 2581-93, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259605

RESUMEN

Metabolic labeling of primate cells revealed the existence of phosphorylated and hypophosphorylated DNA polymerase alpha-primase (Pol-Prim) populations that are distinguishable by monoclonal antibodies. Cell cycle studies showed that the hypophosphorylated form was found in a complex with PP2A and cyclin E-Cdk2 in G1, whereas the phosphorylated enzyme was associated with a cyclin A kinase in S and G2. Modification of Pol-Prim by PP2A and Cdks regulated the interaction with the simian virus 40 origin-binding protein large T antigen and thus initiation of DNA replication. Confocal microscopy demonstrated nuclear colocalization of hypophosphorylated Pol-Prim with MCM2 in S phase nuclei, but its presence preceded 5-bromo-2'-deoxyuridine (BrdU) incorporation. The phosphorylated replicase exclusively colocalized with the BrdU signal, but not with MCM2. Immunoprecipitation experiments proved that only hypophosphorylated Pol-Prim associated with MCM2. The data indicate that the hypophosphorylated enzyme initiates DNA replication at origins, and the phosphorylated form synthesizes the primers for the lagging strand of the replication fork.


Asunto(s)
ADN Primasa/genética , Replicación del ADN , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular , ADN Primasa/inmunología , Humanos , Isoenzimas/genética , Isoenzimas/inmunología , Microscopía Confocal , Fosforilación
4.
Oncogene ; 18(51): 7310-8, 1999 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-10602486

RESUMEN

Transcriptional activation by the tumor suppressor p53 is regulated at multiple levels, including posttranslational modifications of the p53 protein, interaction of p53 with various regulatory proteins, or at the level of sequence-specific DNA binding to the response elements in p53's target genes. We here propose as an additional regulatory mechanism that the DNA topology of p53-responsive promoters may determine the interaction of p53 with its target genes. We demonstrate that sequence-specific DNA binding (SSDB) and transcriptional activation by p53 of the mdm2 promoter is inhibited when this promoter is present in supercoiled DNA, where it forms a non-B-DNA structure which spans the p53-responsive elements. Relaxation of the supercoiled DNA in vitro resulted in conversion of the non-B-DNA to a B-DNA conformation within the mdm2 promoter, and correlated with an enhanced SSDB of p53 and an elevated expression of a reporter gene. In contrast, sequence specific DNA binding and transcriptional activation of the p21 promoter were not inhibited by DNA supercoiling. We propose that conformational alterations within p53-responsive sites, which either promote or prohibit sequence specific DNA binding of p53, are an important feature in orchestrating the activation of different p53 responsive promoters.


Asunto(s)
ADN/genética , Regiones Promotoras Genéticas/genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Sitios de Unión/genética , Línea Celular , ADN/metabolismo , Humanos , Unión Proteica/genética , Análisis de Secuencia de ADN , Proteína p53 Supresora de Tumor/metabolismo
5.
Mol Cell Biol ; 18(9): 5332-42, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9710617

RESUMEN

We demonstrate that wild-type p53 inhibits homologous recombination. To analyze DNA substrate specificities in this process, we designed recombination experiments such that coinfection of simian virus 40 mutant pairs generated heteroduplexes with distinctly unpaired regions. DNA exchanges producing single C-T and A-G mismatches were inhibited four- to sixfold more effectively than DNA exchanges producing G-T and A-C single-base mispairings or unpaired regions of three base pairs comprising G-T/A-C mismatches. p53 bound specifically to three-stranded DNA substrates, mimicking early recombination intermediates. The KD values for the interactions of p53 with three-stranded substrates displaying differently paired and unpaired regions reflected the mismatch base specificities observed in recombination assays in a qualitative and quantitative manner. On the basis of these results, we would like to advance the hypothesis that p53, like classical mismatch repair factors, checks the fidelity of homologous recombination processes by specific mismatch recognition.


Asunto(s)
ADN Viral/metabolismo , Ácidos Nucleicos Heterodúplex/metabolismo , Recombinación Genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Composición de Base , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , ADN Viral/química , Riñón , Macaca mulatta , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Ácidos Nucleicos Heterodúplex/química , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Virus 40 de los Simios/genética , Spodoptera , Especificidad por Sustrato , Transfección , Proteína p53 Supresora de Tumor/biosíntesis
6.
J Virol ; 71(10): 7609-18, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9311842

RESUMEN

Phosphopeptide analyses of the simian virus 40 (SV40) large tumor antigen (LT) in SV40-transformed rat cells, as well as in SV40 lytically infected monkey cells, showed that gel-purified LT that was not complexed to p53 (free LT) and p53-complexed LT differed substantially in their phosphorylation patterns. Most significantly, p53-complexed LT contained phosphopeptides not found in free LT. We show that these additional phosphopeptides were derived from MDM2, a cellular antagonist of p53, which coprecipitated with the p53-LT complexes, probably in a trimeric LT-p53-MDM2 complex. MDM2 also quantitatively bound the free p53 in SV40-transformed cells. Free LT, in contrast, was not found in complex with MDM2, indicating a specific targeting of the MDM2 protein by SV40. This specificity is underscored by significantly different phosphorylation patterns of the MDM2 proteins in normal and SV40-transformed cells. Furthermore, the MDM2 protein, like p53, becomes metabolically stabilized in SV40-transformed cells. This suggests the possibility that the specific targeting of MDM2 by SV40 is aimed at preventing MDM2-directed proteasomal degradation of p53 in SV40-infected and -transformed cells, thereby leading to metabolic stabilization of p53 in these cells.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Transformación Celular Neoplásica , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Virus 40 de los Simios/fisiología , Células 3T3 , Animales , Antígenos Virales de Tumores/química , Antígenos Virales de Tumores/aislamiento & purificación , Línea Celular Transformada , Chlorocebus aethiops , Ratones , Proteínas de Neoplasias/metabolismo , Mapeo Peptídico , Fosfopéptidos/química , Fosfopéptidos/aislamiento & purificación , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-mdm2 , Ratas , Virus 40 de los Simios/patogenicidad , Transcripción Genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/aislamiento & purificación , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...