Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 349
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
HIV Med ; 18(8): 555-563, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28294530

RESUMEN

OBJECTIVES: HIV-associated neurocognitive disorders are highly prevalent, and physical activity (PA) is a modifiable behaviour that may affect neurocognitive function. Our objective was to determine the association between PA and neurocognitive function and the effect of HIV on this association. METHODS: PA was assessed in the Multicenter AIDS Cohort Study with the International Physical Activity Questionnaire. A neuropsychological test battery assessed global impairment and domain-specific impairment (executive function, speed of processing, working memory, learning, memory, and motor function) every 2 years. Semiannually, the Symbol Digit Modalities Test and Trail Making Test Parts A and B were performed. Adjusted logistic regression models were used to assess the PA-neurocognitive function association. Using longitudinal data, we also assessed the PA category-decline of neurocognitive function association with multivariate simple regression. RESULTS: Of 601 men, 44% were HIV-infected. Low, moderate, and high PA was reported in 27%, 25%, and 48% of the HIV-infected men vs. 19%, 32% and 49% of the HIV-uninfected men, respectively. High PA was associated with lower odds of impairment of learning, memory, and motor function [odds ratio (OR) ranging from 0.52 to 0.57; P < 0.05 for all]. The high PA-global impairment association OR was 0.63 [95% confidence interval (CI) 0.39, 1.02]. Among HIV-infected men only, across multiple domains, the high PA-impairment association was even more pronounced (OR from 0.27 to 0.49). Baseline high/moderate PA was not associated with decline of any domain score over time. HIV infection was marginally associated with a higher speed of decline in motor function. CONCLUSIONS: A protective effect of high PA on impairment in neurocognitive domains was observed cross-sectionally. Longitudinal PA measurements are needed to elucidate the PA-neurocognitive function relationship over time.


Asunto(s)
Complejo SIDA Demencia/patología , Cognición , Ejercicio Físico , Infecciones por VIH/complicaciones , Salud Mental , Adulto , Anciano , Anciano de 80 o más Años , Estudios Transversales , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Encuestas y Cuestionarios , Adulto Joven
2.
Nat Genet ; 4(1): 42-6, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8099841

RESUMEN

We report that the expression of murine or human mutant p53 proteins in cells with no endogenous p53 proteins confers new or additional phenotypes upon these cells. Mutant p53 proteins expressed in cell lines lacking p53 resulted in either enhanced tumorigenic potential in nude mice ((10)3 cells) or enhanced plating efficiency in agar cell culture (human SAOS-2 cells). Also, mutant human p53 alleles, unlike the wild-type p53 protein, could also enhance the expression of a test gene regulated by the multi-drug resistance enhancer-promoter element. These data demonstrate a gain of function associated with p53 mutations in addition to the loss of function shown previously to be associated with mutations in this tumour suppressor gene.


Asunto(s)
Genes p53 , Proteína p53 Supresora de Tumor/fisiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , División Celular/genética , Línea Celular , Células Clonales/trasplante , Regulación de la Expresión Génica/genética , Humanos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Neoplasias Experimentales/genética , Fenotipo , Especificidad de la Especie , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
3.
Opt Express ; 19(21): 20571-9, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-21997064

RESUMEN

We used quantitative phase imaging to measure the dispersion relation, i.e. decay rate vs. spatial mode, associated with mass transport in live cells. This approach applies equally well to both discrete and continuous mass distributions without the need for particle tracking. From the quadratic experimental curve specific to diffusion, we extracted the diffusion coefficient as the only fitting parameter. The linear portion of the dispersion relation reveals the deterministic component of the intracellular transport. Our data show a universal behavior where the intracellular transport is diffusive at small scales and deterministic at large scales. Measurements by our method and particle tracking show that, on average, the mass transport in the nucleus is slower than in the cytoplasm.


Asunto(s)
Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Neuroglía/metabolismo , Algoritmos , Animales , Biotecnología/métodos , Difusión , Diseño de Equipo , Hipocampo/metabolismo , Humanos , Luz , Microglía/metabolismo , Microscopía de Interferencia/métodos , Neuronas/metabolismo , Óptica y Fotónica , Dispersión de Radiación , Espectrofotometría/métodos
4.
Nat Med ; 2(7): 804-10, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8673928

RESUMEN

Testicular teratocarcinomas never contain p53 gene mutations even though these tumors express high levels of nuclear p53 protein. We have characterized two murine teratocarcinoma cell lines and find no evidence that endogenous p53-regulated genes are correspondingly upregulated. Differentiation of these teratocarcinoma cells with retinoic acid results in a marked decrease in p53 protein levels but is accompanied by a marked increase in p53-mediated transcriptional activity. Together these results support the hypothesis that the p53 protein in undifferentiated teratocarcinoma cells is transcriptionally inactive and accounts for the lack of selection for p53 gene mutations in this tumor type. These teratocarcinoma cells undergo p53-mediated apoptosis in response to DNA damage, which may explain the routine cures of human testicular tumors with combination chemotherapy.


Asunto(s)
Diferenciación Celular , Daño del ADN , Teratoma/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Apoptosis/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Teratoma/genética , Teratoma/patología , Transcripción Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
5.
J Exp Med ; 188(6): 1017-28, 1998 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-9743520

RESUMEN

A high proportion of tumors arise due to mutation of the p53 tumor suppressor protein. A p53 hotspot mutation at amino acid position 273 from R to H, flanking a peptide epitope that spans residues 264-272, renders cells resistant to killing by human histocompatibility leukocyte antigen (HLA)-A*0201-restricted cytotoxic T lymphocytes (CTLs) specific for this epitope. Acquisition of the R to H mutation at residue 273 of the human p53 protein promotes tumor growth in vivo by selective escape from recognition by p53.264-272 peptide-specific CTLs. Synthetic 27-mer p53 polypeptides covering the antigenic nonamer region 264-272 of p53 were used as proteasome substrates to investigate whether the R to H mutation at the P1' position of the COOH terminus of the epitope affects proteasome-mediated processing of the protein. Analysis of the generated products by tandem mass spectrometry and the kinetics of polypeptide processing in conjunction with CTL assays demonstrate that the R to H mutation alters proteasomal processing of the p53 protein by inhibiting proteolytic cleavage between residues 272 and 273. This prevents the release of the natural CTL epitope that spans flanking residues 264-272 as well as a putative precursor peptide. These results demonstrate that mutation of p53 not only leads to malignant transformation but may also, in some instances, affect immune surveillance and should be considered in the design of cancer vaccines.


Asunto(s)
Citotoxicidad Inmunológica/genética , Epítopos de Linfocito T/inmunología , Mutación Puntual/inmunología , Linfocitos T Citotóxicos/inmunología , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/genética , Arginina/genética , Sitios de Unión/genética , Sitios de Unión/inmunología , División Celular/genética , División Celular/inmunología , Cisteína Endopeptidasas/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Histidina/genética , Humanos , Hidrólisis , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Complejos Multienzimáticos/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Péptidos/síntesis química , Péptidos/inmunología , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal , Especificidad por Sustrato/genética , Especificidad por Sustrato/inmunología , Células Tumorales Cultivadas
6.
Science ; 248(4951): 76-9, 1990 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-2157286

RESUMEN

Human papillomavirus type 16 (HPV-16) is a DNA tumor virus that is associated with human anogenital cancers and encodes two transforming proteins, E6 and E7. The E7 protein has been shown to bind to the retinoblastoma tumor suppressor gene product, pRB. This study shows that the E6 protein of HPV-16 is capable of binding to the cellular p53 protein. The ability of the E6 proteins from different human papillomaviruses to form complexes with p53 was assayed and found to correlate with the in vivo clinical behavior and the in vitro transforming activity of these different papillomaviruses. The wild-type p53 protein has tumor suppressor properties and has also been found in association with large T antigen and the E1B 55-kilodalton protein in cells transformed by SV40 and by adenovirus type 5, respectively, providing further evidence that the human papillomaviruses, the adenoviruses, and SV40 may effect similar cellular pathways in transformation.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Oncogénicas Virales/metabolismo , Proteínas Oncogénicas/metabolismo , Papillomaviridae/análisis , Fosfoproteínas/metabolismo , Proteínas Precoces de Adenovirus , Secuencia de Aminoácidos , Antígenos Transformadores de Poliomavirus/metabolismo , Secuencia de Bases , Transformación Celular Neoplásica , Transformación Celular Viral , Clonación Molecular , Humanos , Técnicas de Inmunoadsorción , Datos de Secuencia Molecular , Mutación , Proteínas Oncogénicas/genética , Proteínas Oncogénicas Virales/genética , Fosfoproteínas/genética , Reacción en Cadena de la Polimerasa , Unión Proteica , Virus 40 de los Simios/inmunología , Proteína p53 Supresora de Tumor
7.
Science ; 172(3990): 1345-6, 1971 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-4325520

RESUMEN

The change in the surface structure of cultutred mammalian cells infected with oncogenic DNA viruses was similar to that described for the fully and permanently transformed cell surfaces. The course of appearance of this change was established. Synthesis of host DNA is required for expression of the surface change triggered by infection with the oncogenic virus.


Asunto(s)
Adenoviridae/metabolismo , Transformación Celular Neoplásica , ADN de Neoplasias/biosíntesis , Virus Oncogénicos/metabolismo , Virus 40 de los Simios/metabolismo , Propiedades de Superficie , Animales , Línea Celular/metabolismo , Línea Celular/microbiología , Haplorrinos , Riñón , Lectinas/farmacología
8.
Science ; 277(5330): 1310-3, 1997 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-9271577

RESUMEN

Activation domains are functional modules that enable sequence-specific DNA binding proteins to stimulate transcription. The structural basis for the function of activation domains is poorly understood. A combination of nuclear magnetic resonance (NMR) and biochemical experiments revealed that the minimal acidic activation domain of the herpes simplex virus VP16 protein undergoes an induced transition from random coil to alpha helix upon binding to its target protein, hTAFII31 (a human TFIID TATA box-binding protein-associated factor). Identification of the two hydrophobic residues that make nonpolar contacts suggests a general recognition motif of acidic activation domains for hTAFII31.


Asunto(s)
Proteína Vmw65 de Virus del Herpes Simple/química , Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Estructura Secundaria de Proteína , Factores Asociados con la Proteína de Unión a TATA , Transactivadores/metabolismo , Factor de Transcripción TFIID , Activación Transcripcional , Secuencia de Aminoácidos , Humanos , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Pliegue de Proteína , Eliminación de Secuencia , Transactivadores/química
9.
Science ; 274(5289): 948-53, 1996 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-8875929

RESUMEN

The MDM2 oncoprotein is a cellular inhibitor of the p53 tumor suppressor in that it can bind the transactivation domain of p53 and downregulate its ability to activate transcription. In certain cancers, MDM2 amplification is a common event and contributes to the inactivation of p53. The crystal structure of the 109-residue amino-terminal domain of MDM2 bound to a 15-residue transactivation domain peptide of p53 revealed that MDM2 has a deep hydrophobic cleft on which the p53 peptide binds as an amphipathic alpha helix. The interface relies on the steric complementarity between the MDM2 cleft and the hydrophobic face of the p53 alpha helix and, in particular, on a triad of p53 amino acids-Phe19, Trp23, and Leu26-which insert deep into the MDM2 cleft. These same p53 residues are also involved in transactivation, supporting the hypothesis that MDM2 inactivates p53 by concealing its transactivation domain. The structure also suggests that the amphipathic alpha helix may be a common structural motif in the binding of a diverse family of transactivation factors to the TATA-binding protein-associated factors.


Asunto(s)
Proteínas Nucleares , Conformación Proteica , Proteínas Proto-Oncogénicas/química , Activación Transcripcional , Proteína p53 Supresora de Tumor/química , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
10.
Science ; 291(5507): 1304-51, 2001 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-11181995

RESUMEN

A 2.91-billion base pair (bp) consensus sequence of the euchromatic portion of the human genome was generated by the whole-genome shotgun sequencing method. The 14.8-billion bp DNA sequence was generated over 9 months from 27,271,853 high-quality sequence reads (5.11-fold coverage of the genome) from both ends of plasmid clones made from the DNA of five individuals. Two assembly strategies-a whole-genome assembly and a regional chromosome assembly-were used, each combining sequence data from Celera and the publicly funded genome effort. The public data were shredded into 550-bp segments to create a 2.9-fold coverage of those genome regions that had been sequenced, without including biases inherent in the cloning and assembly procedure used by the publicly funded group. This brought the effective coverage in the assemblies to eightfold, reducing the number and size of gaps in the final assembly over what would be obtained with 5.11-fold coverage. The two assembly strategies yielded very similar results that largely agree with independent mapping data. The assemblies effectively cover the euchromatic regions of the human chromosomes. More than 90% of the genome is in scaffold assemblies of 100,000 bp or more, and 25% of the genome is in scaffolds of 10 million bp or larger. Analysis of the genome sequence revealed 26,588 protein-encoding transcripts for which there was strong corroborating evidence and an additional approximately 12,000 computationally derived genes with mouse matches or other weak supporting evidence. Although gene-dense clusters are obvious, almost half the genes are dispersed in low G+C sequence separated by large tracts of apparently noncoding sequence. Only 1.1% of the genome is spanned by exons, whereas 24% is in introns, with 75% of the genome being intergenic DNA. Duplications of segmental blocks, ranging in size up to chromosomal lengths, are abundant throughout the genome and reveal a complex evolutionary history. Comparative genomic analysis indicates vertebrate expansions of genes associated with neuronal function, with tissue-specific developmental regulation, and with the hemostasis and immune systems. DNA sequence comparisons between the consensus sequence and publicly funded genome data provided locations of 2.1 million single-nucleotide polymorphisms (SNPs). A random pair of human haploid genomes differed at a rate of 1 bp per 1250 on average, but there was marked heterogeneity in the level of polymorphism across the genome. Less than 1% of all SNPs resulted in variation in proteins, but the task of determining which SNPs have functional consequences remains an open challenge.


Asunto(s)
Genoma Humano , Proyecto Genoma Humano , Análisis de Secuencia de ADN , Algoritmos , Animales , Bandeo Cromosómico , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Biología Computacional , Secuencia de Consenso , Islas de CpG , ADN Intergénico , Bases de Datos Factuales , Evolución Molecular , Exones , Femenino , Duplicación de Gen , Genes , Variación Genética , Humanos , Intrones , Masculino , Fenotipo , Mapeo Físico de Cromosoma , Polimorfismo de Nucleótido Simple , Proteínas/genética , Proteínas/fisiología , Seudogenes , Secuencias Repetitivas de Ácidos Nucleicos , Retroelementos , Análisis de Secuencia de ADN/métodos , Especificidad de la Especie
11.
Oncogene ; 26(9): 1317-23, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17322917

RESUMEN

Cancer biology finds itself in a post-genomic era and the hopes of using inherited genetic variants to improve prevention and treatment strategies are widespread. One of the largest types of inherited genetic variation is the single nucleotide polymorphism (SNP), of which there are at least 4.5 million. The challenge now becomes how to discover which polymorphisms alter cancer in humans and how to begin to understand their mechanism of action. In this report, a series of recent publications will be reviewed that have studied a polymorphism in the p53 tumor suppressor pathway, MDM2 SNP309. These reports have lent insights into how germline genetic variants of the p53 pathway could interact with gender, environmental stresses and tumor genetics to affect cancer in humans. Importantly, these observations have also exposed potential nodes of intervention, which could prove valuable in both the prevention and treatment of this disease in humans.


Asunto(s)
Neoplasias/genética , Estrés Oxidativo , Polimorfismo de Nucleótido Simple , Factores Sexuales , Proteína p53 Supresora de Tumor/genética , Estrógenos/metabolismo , Femenino , Humanos , Masculino , Mutación , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Fumar , Proteína p53 Supresora de Tumor/metabolismo , Virosis
12.
Curr Opin Genet Dev ; 3(1): 50-4, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8453275

RESUMEN

The p53 tumor suppressor is a transcription factor that can activate the expression of some genes and repress the transcription of others. The protein appears to be dispensable for normal murine development, although mice lacking p53 develop tumors at an early age and their fibroblasts are genetically unstable in culture. Human and murine cells lacking wild-type p53 loose the ability to arrest in the G1 phase of the cell cycle in response to gamma-irradiation. Therefore, p53 may be a cell-cycle checkpoint protein that regulates the cycle under adverse conditions.


Asunto(s)
Ciclo Celular , Proteína p53 Supresora de Tumor/fisiología , Animales , Supervivencia Celular , Daño del ADN , Regulación de la Expresión Génica , Genes p53 , Humanos , Mutación , Neoplasias/genética , Virus Oncogénicos/genética , Proteínas Represoras/fisiología , Transactivadores/fisiología , Transcripción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
13.
Oncogene ; 25(1): 1-7, 2006 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-16278683

RESUMEN

The p53 protein is a transcription factor that integrates various cellular stress signals. The accumulation of the mutant huntingtin protein with an expanded polyglutamine tract plays a central role in the pathology of human Huntington's disease. We found that the huntingtin gene contains multiple putative p53-responsive elements and p53 binds to these elements both in vivo and in vitro. p53 activation in cultured human cells, either by a temperature-sensitive mutant p53 protein or by gamma-irradiation (gamma-irradiation), increases huntingtin mRNA and protein expression. Similarly, murine huntingtin also contains multiple putative p53-responsive elements and its expression is induced by p53 activation in cultured cells. Moreover, gamma-irradiation, which activates p53, increases huntingtin gene expression in the striatum and cortex of mouse brain, the major pathological sites for Huntington's disease, in p53+/+ but not the isogenic p53-/- mice. These results demonstrate that p53 protein can regulate huntingtin expression at transcriptional level, and suggest that a p53 stress response could be a modulator of the process of Huntington's disease.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Proteína p53 Supresora de Tumor/fisiología , Animales , Northern Blotting , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Daño del ADN , Rayos gamma , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , ARN Mensajero/metabolismo , Elementos de Respuesta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Temperatura , Factores de Tiempo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Cell Death Differ ; 13(6): 1027-36, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16557269

RESUMEN

The p53 pathway is composed of hundreds of genes and their products that respond to a wide variety of stress signals. These responses to stress include apoptosis, cellular senescence or cell cycle arrest. In addition the p53-regulated genes produce proteins that communicate these stress signals to adjacent cells, prevent and repair damaged DNA and create feedback loops that enhance or attenuate p53 activity and communicate with other signal transduction pathways. Many questions remain to be explored in our understanding of how this network of genes plays a role in protection from cancers, therapy and integrating the homeostatic mechanisms of stress management and fidelity in a cell and organism. The goal of this chapter is to elucidate some of those questions and suggest new directions for this area of research.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , ADN/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , ADN/genética , Humanos , Mutación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Elementos de Respuesta/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
15.
Mol Cell Biol ; 1(2): 101-10, 1981 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-6100960

RESUMEN

The 54K cellular tumor antigen has been translated in vitro, using messenger ribonucleic acids from simian virus 40 (SV40)-transformed cells or 3T3 cells. The in vitro 54K product could be immunoprecipitated with SV40 tumor serum and had a peptide map that was similar, but not identical, to the in vivo product. The levels of this 54K protein in SV3T3 cells were significantly higher than those detected in 3T3 cells (D. I. H. Linzer, W. Maltzman, and A. J. Levine, Virology 98:308-318, 1979). In spite of this, the levels of translatable 54K messenger ribonucleic acid from 3T3 and SV3T3 cells were roughly equivalent or often greater in 3T3 cells. Pulse-chase experiments with the 54K protein from 3T3 or SV3T3 cells demonstrated that this protein, once synthesized, was rapidly degraded in 3T3 cells but was extremely stable in SV3T3 cells. Similarly, in an SV40 tsA-transformed cell line, temperature sensitive for the SV40 T-antigen, the 54K protein was rapidly turned over at the nonpermissive temperature and stable at the permissive temperature, whereas the levels of translatable 54K messenger ribonucleic acid at each temperature were roughly equal. These results demonstrate a post-translational regulation of the 54K cellular tumor antigen and suggest that this control is mediated by the SV40 large T-antigen.


Asunto(s)
Antígenos Virales de Tumores/genética , Biosíntesis de Proteínas , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Línea Celular Transformada , ARN Mensajero/genética , ARN Mensajero/metabolismo , Virus 40 de los Simios/genética , Virus 40 de los Simios/inmunología
16.
Mol Cell Biol ; 13(7): 4107-14, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7686617

RESUMEN

The 90-kDa cellular protein encoded by the mouse mdm-2 oncogene binds to the p53 protein in vivo and inhibits its transactivation function (J. Momand, G. P. Zambetti, D. C. Olson, D. George, and A. J. Levine, Cell 69:1237-1245, 1992). cDNA clones encoding the human homolog of the mdm-2 protein (also called hdm-2) were isolated from a HeLa cell cDNA library. A series of monoclonal antibodies have been generated against human mdm-2 protein, and the epitopes recognized by these antibodies have been mapped. By construction of a series of deletion mutants, the region of the mdm-2 protein that is critical for complex formation with the p53 protein has been mapped to the N-terminal portion of the human mdm-2 protein. Interestingly, a monoclonal antibody with an epitope located in this same region failed to immunoprecipitate the mdm-2-p53 complex and appeared to recognize only free mdm-2 protein. The domain of the p53 protein that is sufficient for interaction with human mdm-2 protein has been mapped to the N-terminal 52 amino acid residues of the p53 protein. This region contains the transactivation domain of p53, suggesting that mdm-2 may inhibit p53 function by disrupting its interaction with the general transcription machinery.


Asunto(s)
Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor/metabolismo , Animales , Anticuerpos Monoclonales , Secuencia de Bases , Sitios de Unión , ADN/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Epítopos , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Oncogenes , Proteínas Proto-Oncogénicas c-mdm2 , Proteína p53 Supresora de Tumor/genética
17.
Mol Cell Biol ; 5(10): 2533-42, 1985 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-3016506

RESUMEN

The Epstein-Barr virus (EBV) genome becomes established as a multicopy plasmid in the nucleus of infected B lymphocytes. A cis-acting DNA sequence previously described within the BamHI-C fragment of the EBV genome (J. Yates, N. Warren, D. Reisman, and B. Sugden, Proc. Natl. Acad. Sci. USA 81:3806-3810, 1984) allows stable extrachromosomal plasmid maintenance in latently infected cells, but not in EBV-negative cells. In agreement with the findings of Yates et al., deletion analysis permitted the assignment of this function to a 2,208-base-pair region (nucleotides 7315 to 9517 of the B95-8 strain of EBV) of the BamHI-C fragment that contained a striking repetitive sequence and an extended region of dyad symmetry. A recombinant vector, p410+, was constructed which carried the BamHI-K fragment (nucleotides 107565 to 112625 of the B95-8 strain, encoding the EBV-associated nuclear antigen EBNA-1), the cis-acting sequence from the BamHI-C fragment, and a dominant selectable marker gene encoding G-418 resistance in animal cells. After being transfected into HeLa cells, this plasmid persisted extrachromosomally at a low copy number, with no detectable rearrangements or deletions. Two mutations in the BamHI-K-derived portion of p410+, a large in-frame deletion and a linker insertion frameshift mutation, both of which alter the carboxy-terminal portion of EBNA-1, destroyed the ability of the plasmid to persist extrachromosomally in HeLa cells. A small in-frame deletion and linker insertion mutation in the region encoding the carboxy-terminal portion of EBNA-1, which replaced 19 amino acid codons with 2, had no effect on the maintenance of p410+ in HeLa cells. These observations indicate that EBNA-1, in combination with a cis-acting sequence in the BamHI-C fragment, is in part responsible for extrachromosomal EBV-derived plasmid maintenance in HeLa cells. Two additional activities have been localized to the BamHI-C DNA fragment: (i) a DNA sequence that could functionally substitute for the simian virus 40 enhancer and promoter elements controlling the expression of G-418 resistance and (ii) a DNA sequence which, although not sufficient to allow extrachromosomal plasmid maintenance, enhanced the frequency of transformation to G-418 resistance in EBV-positive (but not EBV-negative) cells. These findings suggest that the BamHI-C fragment contains a lymphoid-specific or EBV-inducible promoter or enhancer element or both.


Asunto(s)
ADN Viral/genética , Herpesvirus Humano 4/genética , Antígenos Virales/genética , Mapeo Cromosómico , Enzimas de Restricción del ADN , Elementos de Facilitación Genéticos , Antígenos Nucleares del Virus de Epstein-Barr , Herencia Extracromosómica , Genes Virales , Células HeLa , Humanos , Plásmidos , Regiones Promotoras Genéticas , Replicón
18.
Mol Cell Biol ; 18(12): 7288-93, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9819415

RESUMEN

The MDM2 oncoprotein targets the p53 tumor suppressor protein for degradation when the two proteins are expressed in cells. The regulation of p53 levels by MDM2 requires the ability of MDM2 to be exported from the nucleus by utilizing its nuclear export signal (NES). The drug leptomycin B (LMB) blocks the formation of nuclear export complexes consisting of CRM1, RanGTP, and NES-containing proteins. It is predicted that LMB should inhibit nuclear-cytoplasmic shuttling by MDM2 and subsequently stabilize p53. This communication demonstrates that LMB treatment of various cell lines led to an increase in the steady-state levels of the p53 protein as a result of an increase in its stability. The stabilized p53 protein localized to the nucleus and was an active transcription factor. These results indicate that the low steady-state levels of p53 in the absence of DNA damage result from p53's nuclear export for cytoplasmic degradation. LMB also led to p53 stabilization in cell lines that contain human papillomavirus (HPV) DNA and express HPV E6, a protein that targets p53 for degradation. MDM2 is not necessary for E6-dependent degradation of p53, as evidenced by the observation that E6 promoted p53 degradation in cells lacking endogenous MDM2. In addition, LMB reduced E6's ability to degrade p53 in the absence of MDM2, demonstrating that complete degradation of p53 by E6 requires nuclear export and therefore likely occurs in cytoplasmic proteasomes. These data suggest that the nuclear export of p53 to the cytoplasm for degradation is a general mechanism for regulating p53 levels.


Asunto(s)
Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Nucleares , Papillomaviridae/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Ácidos Grasos Insaturados/farmacología , Humanos , Inmunohistoquímica , Proteínas Proto-Oncogénicas c-mdm2 , Transcripción Genética/genética , Células Tumorales Cultivadas
19.
Mol Cell Biol ; 21(2): 562-74, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134343

RESUMEN

The Wnt/Wg signaling pathway functions during development to regulate cell fate determination and patterning in various organisms. Two pathways are reported to lie downstream of Wnt signaling in vertebrates. The canonical pathway relies on the activation of target genes through the beta-catenin-Lef/TCF complex, while the noncanonical pathway employs the activation of protein kinase C (PKC) and increases in intracellular calcium to induce target gene expression. cDNA subtractive hybridization between a cell line that overexpresses Wnt-1 (C57MG/Wnt-1) and the parental cell line (C57MG) was performed to identify downstream target genes of Wnt-1 signaling. Among the putative Wnt-1 target genes, we have identified a mouse homolog of the gene encoding human transcription factor basic transcription element binding protein 2 (mBTEB2). The mBTEB2 transcript is found at high levels in mammary tissue taken from a transgenic mouse overexpressing Wnt-1 (both tissue prior to active proliferation and tumor tissue) but is barely detectable in wild-type mouse mammary glands. The regulation of mBTEB2 by Wnt-1 signaling in tissue culture occurs through a beta-catenin-Lef/TCF-independent mechanism, as it is instead partially regulated by PKC. The Wnt-1-induced, PKC-dependent activation of mouse BTEB2 in C57MG cells, as well as the ability of Wnt-1 to stabilize beta-catenin in these cells, is consistent with the hypothesis that both the noncanonical and canonical Wnt pathways are activated concomitantly in the same cell. These results suggest that mBTEB2 is a biologically relevant target of Wnt-1 signaling that is activated through a beta-catenin-independent, PKC-sensitive pathway in response to Wnt-1.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Transactivadores/genética , Proteínas de Pez Cebra , Animales , Línea Celular , Técnicas de Cocultivo , Activación Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Respuesta/genética , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Transactivadores/metabolismo , Proteínas Wnt , Proteína Wnt1 , beta Catenina
20.
Mol Cell Biol ; 16(4): 1786-93, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8657154

RESUMEN

The p21WAF-1 gene is positively regulated by the wild-type p53 protein. p21WAF-1 has been shown to interact with several cyclin-dependent kinase complexes and block the activity of G1 cyclin-dependent kinases (cdks). Mutational analysis with the p21WAF-1 gene localized a site, at amino acid residues 21 and 24 in the amino terminus of the protein, for p21WAF-1 binding to cyclins D and E. This region of the protein is conserved (residues 21 to 26) in other p21WAF-1 family members, p27kip-1 and p57kip-2. The same p21WAF-121,24 mutant also fails to bind to cyclin D1-cdk 4 or cyclin E-cdk 2 complexes in vitro, suggesting that amino acid residues 21 and 24 are important for p21WAF-1-cdk-cyclin trimeric complex interactions. The p21WAF-1 wild-type protein will suppress tumor cell growth in culture while p21WAF-1 mutant proteins with defects in residues 21 and 24 fail to suppress tumor cell growth. The overexpression of cyclin D or E in these cells will partially overcome the growth suppression of wild-type p21WAF-1 protein in cells. These results provide evidence that p21WAF-1 acts through cyclin D1-cdk4 and cyclin E-cdk2 complexes in vivo to induce the growth suppression. The p21WAF-1 binding sites for cyclins (residues 21 to 26), cdk2 (residues 49 to 71), and proliferating-cell nuclear antigen (residues 124 to 164) have all been mapped to discrete sites on the protein.


Asunto(s)
Ciclinas/genética , Secuencia de Aminoácidos , División Celular/genética , Ciclina D , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Genes p53 , Humanos , Datos de Secuencia Molecular , Mutación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA