Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-33868793

RESUMEN

A novel system to cultivate and record brain slices directly on high-density microelectrode arrays (HD-MEA) was developed. This system allows to continuously record electrical activity of selected individual neurons at high spatial resolution, while monitoring neuronal network activity at the same time. For the first time, properties of single neurons and the corresponding neuronal network in an organotypic hippocampal slice culture were studied over four consecutive weeks at daily intervals.

2.
Int J Oncol ; 28(2): 421-30, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16391797

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in a variety of tumor cells through two of its receptors: TRAIL-R1 and TRAIL-R2. We investigate the susceptibility of human renal cell carcinoma (RCC) cells to TRM-1 and HGS-ETR2, 2 human monoclonal agonistic antibodies specific for TRAIL-R1 and TRAIL-R2, respectively. HGS-ETR2 effectively induced apoptotic cell death in 10 of 11 cell cultures, including 2 human RCC cell lines and 9 human primary RCC cell cultures, with a more pronounced effect after preincubation with anti-human IgG Fc. In contrast, TRM-1 was effective in only 1 primary RCC cell culture. The increased effectiveness of HGS-ETR2 for inducing cell death might have been affected by differences in the cell-surface expression of the 2 TRAIL receptors, namely that TRAIL-R2 but not TRAIL-R1 was frequently expressed in most of the RCC cells tested. The activities of caspase-9, -8, -6, and -3 were increased with HGS-ETR2-induced apoptosis, and cell death could be blocked by specific caspase inhibitors for caspase-9, -8, and -3, and the general caspase inhibitor. In vivo administration of HGS-ETR2 with or without cross-linker significantly suppressed tumor growth of subcutaneously inoculated human RCC xenografts in immunodeficient mice. These results suggest the potential utility of TRAIL-R2 antibody as a novel therapeutic agent in RCC.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoptosis , Carcinoma de Células Renales/prevención & control , Neoplasias Renales/prevención & control , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis/inmunología , Carcinoma de Células Renales/patología , Inhibidores de Caspasas , Caspasas/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias Renales/patología , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Br J Cancer ; 92(8): 1430-41, 2005 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-15846298

RESUMEN

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a variety of tumour cells through activation of TRAIL-R1 and TRAIL-R2 death signalling receptors. Here, we describe the characterisation and activity of HGS-ETR1, the first fully human, agonistic TRAIL-R1 mAb that is being developed as an antitumour therapeutic agent. HGS-ETR1 showed specific binding to TRAIL-R1 receptor. HGS-ETR1 reduced the viability of multiple types of tumour cells in vitro, and induced activation of caspase 8, Bid, caspase 9, caspase 3, and cleavage of PARP, indicating activation of TRAIL-R1 alone was sufficient to induce both extrinsic and intrinsic apoptotic pathways. Treatment of cell lines in vitro with HGS-ETR1 enhanced the cytotoxicity of chemotherapeutic agents (camptothecin, cisplatin, carboplatin, or 5-fluorouracil) even in tumour cell lines that were not sensitive to HGS-ETR1 alone. In vivo administration of HGS-ETR1 resulted in rapid tumour regression or repression of tumour growth in pre-established colon, non-small-cell lung, and renal tumours in xenograft models. Combination of HGS-ETR1 with chemotherapeutic agents (topotecan, 5-fluorouracil, and irinotecan) in three independent colon cancer xenograft models resulted in an enhanced antitumour efficacy compared to either agent alone. Pharmacokinetic studies in the mouse following intravenous injection showed that HGS-ETR1 serum concentrations were biphasic with a terminal half-life of 6.9-8.7 days and a steady-state volume of distribution of approximately 60 ml kg(-1). Clearance was 3.6-5.7 ml(-1) day(-1) kg(-1). These data suggest that HGS-ETR1 is a specific and potent antitumour agent with favourable pharmacokinetic characteristics and the potential to provide therapeutic benefit for a broad range of human malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Antineoplásicos/inmunología , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Semivida , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF
4.
Oncogene ; 17(21): 2691-700, 1998 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-9840933

RESUMEN

The Met tyrosine kinase receptor has been implicated in human cancer. Here we have examined the signaling requirements of three oncogenic forms of this molecule: wild type Met in response to ligand/autocrine stimulation, Met which has been mutationally activated, and Tpr-Met (a constitutively active truncated Met fusion protein). Previous studies have demonstrated the importance of a Grb2 binding site, and of specific tyrosine residues (i.e. Y8,9 and Y14,15) for Met function, and we have now explored the relevance of these and other sites for oncogenic Met signaling. Following substitution of various intracellular tyrosines for phenylalanine, we find that the transforming activity of each Met oncogene is dependent upon tyrosines Y8,9 and Y14,15, in addition to two novel tyrosines (Y6 and Y10) not previously implicated in Met signaling. Tyrosines Y6 and Y10 influence a variety of Met-mediated responses both in vitro (transformation, mitogenicity and invasion), and in vivo (tumorigenicity and metastasis). We also show that Tpr-Met is much more dependent on its Grb2 binding site for biological activity than are the other oncogenic forms of the Met receptor. Thus, although the three Met oncogenes examined are similar in their dependency on a number of specific tyrosines for activity, the signaling strategy employed by Tpr-Met can be differentiated from that of the other two.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Sustitución de Aminoácidos , Transformación Celular Neoplásica/genética , Oncogenes , Proteínas Proto-Oncogénicas c-met/fisiología , Transducción de Señal , Células 3T3 , Animales , Sitios de Unión , Femenino , Proteína Adaptadora GRB2 , Ratones , Ratones Desnudos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Metástasis de la Neoplasia , Fenilalanina/química , Mutación Puntual , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Recombinantes de Fusión/fisiología , Eliminación de Secuencia , Relación Estructura-Actividad , Tirosina/química
5.
Oncogene ; 17(16): 2019-25, 1998 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-9798673

RESUMEN

Aberrations in Met-hepatocyte growth factor/scatter factor (HGF/SF) signaling have been implicated in the acquisition of tumorigenic and metastatic phenotypes. Here we show that murine NIH3T3 and C127 cells transformed by the Ras oncogene overexpress the Met receptor, resulting in enhanced HGF/SF-mediated responses in vitro including invasion through basement membrane. Accompanying the increase in Met in ras-transformed NIH3T3 cells, there is a decrease in endogenous HGF/SF expression as previously observed in cells exogenously overexpressing Met. However, subcutaneously grown tumors and experimental lung metastases derived from these cells express significantly higher levels of endogenous HGF/SF together with high levels of Met. These results suggest Met-HGF/SF signaling enhances tumor growth and metastasis of Ras-transformed NIH3T3 cells.


Asunto(s)
Transformación Celular Neoplásica , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , Células 3T3 , Animales , Línea Celular , Línea Celular Transformada , Perros , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Ratones Desnudos , Proteína Oncogénica p21(ras)/genética , Proteínas Proto-Oncogénicas c-met/biosíntesis
6.
Proc Natl Acad Sci U S A ; 95(24): 14417-22, 1998 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9826715

RESUMEN

Mutations in Met have been identified in human papillary renal carcinomas. We have shown previously that these mutations deregulate the enzymatic activity of Met and that NIH 3T3 cells expressing mutationally activated Met are transformed in vitro and are tumorigenic in vivo. In the present investigation, we find that mutant Met induces the motility of Madin-Darby canine kidney cells in vitro and experimental metastasis of NIH 3T3 cells in vivo, and that the Ras-Raf-MEK-ERK signaling pathway, which has been implicated previously in cellular motility and metastasis, is constitutively activated by the Met mutants. We also report that transgenic mice harboring mutationally activated Met develop metastatic mammary carcinoma. These data confirm the tumorigenic activity of mutant Met molecules and demonstrate their ability to induce the metastatic phenotype.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Mamarias Experimentales/patología , Proteínas Proto-Oncogénicas c-met/genética , Células 3T3 , Animales , Línea Celular , Movimiento Celular/fisiología , Perros , Femenino , Humanos , Riñón , Neoplasias Mamarias Experimentales/genética , Metalotioneína/biosíntesis , Metalotioneína/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Metástasis de la Neoplasia , Mutación Puntual , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Transfección
7.
Proc Natl Acad Sci U S A ; 94(12): 6048-53, 1997 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-9177166

RESUMEN

Exposure of mammalian cells to ionizing radiation (IR) induces a complex array of cellular responses including cell cycle arrest and/or apoptosis. IR-induced G1 arrest has been shown to depend on the presence of the tumor suppressor p53, which acts as a transcriptional activator of several genes. p53 also plays a role in the induction of apoptosis in response to DNA damage, and this pathway can be activated by both transcription-dependent and -independent mechanisms. Here we report the identification of a novel transcript whose expression is induced in response to IR in a p53-dependent manner, and that shows homology to the type 2C protein phosphatases. We have named this novel gene, wip1. In vitro, recombinant Wip1 displayed characteristics of a type 2C phosphatase, including Mg2+ dependence and relative insensitivity to okadaic acid. Studies performed in several cell lines revealed that wip1 accumulation following IR correlates with the presence of wild-type p53. The accumulation of wip1 mRNA following IR was rapid and transient, and the protein was localized to the nucleus. Similar to waf1, ectopic expression of wip1 in human cells suppressed colony formation. These results suggest that Wip1 might contribute to growth inhibitory pathways activated in response to DNA damage in a p53-dependent manner.


Asunto(s)
Fosfoproteínas Fosfatasas/biosíntesis , Proteínas Tirosina Fosfatasas/biosíntesis , Transcripción Genética/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Ciclo Celular , Línea Celular , Clonación Molecular , Secuencia de Consenso , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Inducción Enzimática/efectos de la radiación , Inhibidores Enzimáticos , Fase G1 , Humanos , Cinética , Ratones , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/efectos de la radiación , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/efectos de la radiación , ARN Mensajero/biosíntesis , Radiación Ionizante , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de la radiación , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Células Tumorales Cultivadas
8.
Hum Immunol ; 53(1): 23-33, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9127144

RESUMEN

A beta 2-microglobulin (beta 2m)-deficient kidney carcinoma cell line and three monoclonal antibodies to the alpha 1 (L31), alpha 2 (W6/32), and alpha 3 (Q1/28) domain of class I HLA molecules were selected to assess the role of beta 2m in regulating the conformation and surface expression of HLA-C molecules. HLA-A2, -B27, and -CW1 molecules synthesized by beta 2m-deficient cells were compared to heavy chains synthesized in transfectants expressing a large excess of beta 2m. As assessed by differential binding with monoclonal antibodies and partitioning studies in the detergent TX-114, no HLA-A2, -B27, or -CW1 molecules can be expressed, in a correct conformation, by beta 2m-deficient cells. These cells, however, do express low but significant amounts of free HLA-CW1 heavy chains at the cell surface. Transfection with beta 2m causes a coordinate change in the antibody reactivity of the three domains of HLA-CW1 molecules, thereby providing the first experimental demonstration that assembly with beta 2m affects the folding of not only the alpha 1 and alpha 2, but also of the alpha 3 domain. HLA-CW1 heavy chains, when free of beta 2m, are less soluble in the detergent TX-114 than free HLA-B27 heavy chains, and when associated with beta 2m share an alpha 3 domain epitope with free HLA-A2 and -B27 heavy chains. Moreover, their assembly with beta 2m is largely incomplete. Those data additionally demonstrate an impaired ability of HLA-CW1 to properly fold and establish a close similarity of HLA-CW1 to murine Db and Ld molecules. Although the functional role, if any, of free HLA-CW1 heavy chains remains to be determined, the present study demonstrates that the absence of beta 2m does not completely ablate class I expression in neoplastic cells of human origin.


Asunto(s)
Antígenos HLA-C/biosíntesis , Antígenos HLA-C/química , Conformación Proteica , Microglobulina beta-2/deficiencia , Carcinoma de Células Renales , Separación Celular , Detergentes , Genotipo , Antígenos HLA-A/genética , Antígenos HLA-B/genética , Antígenos HLA-C/genética , Humanos , Focalización Isoeléctrica , Neoplasias Renales , Octoxinol , Polietilenglicoles , Transfección , Células Tumorales Cultivadas , Microglobulina beta-2/genética
9.
J Biol Chem ; 270(49): 29386-91, 1995 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-7493974

RESUMEN

Incubating human cells in diethylmaleate (DEM) depletes the intracellular pool of reduced glutathione (GSH) and increases the concentration of oxidative free radicals. We found that DEM-induced oxidative stress reduced the ability of p53 to bind its consensus recognition sequence and to activate transcription of a p53-specific reporter gene. Nevertheless, DEM treatment induced expression of WAF1/CIP1 but not GADD45 mRNA. The fact that N-acetylcysteine, a precursor of GSH that blocks oxidative stress, prevented WAF1/CIP1 induction by DEM suggests that WAF1/CIP1 induction probably was a consequence of the ability of DEM to reduce intracellular GSH levels. DEM induced WAF1/CIP1 expression in Saos-2 and T98G cells, both of which lack functional p53 protein. DEM treatment did not produce an increase in membrane-associated protein kinase C, but ERK2, a mitogen-activated protein kinase, was phosphorylated in a manner consistent with ERK2 activation. DEM treatment also produced a dose-dependent delay in cell cycle progression, which at low concentrations (0.25 mM) consisted of a G2/M arrest and at higher concentrations (1 mM) also involved G1 and S phase delays. Our results indicate that oxidative stress induces WAF1/CIP1 expression and arrests cell cycle progression through a mechanism that is independent of p53. This mechanism may provide for cell cycle checkpoint control under conditions that inactivate p53.


Asunto(s)
Ciclinas/biosíntesis , Inhibidores Enzimáticos , Estrés Oxidativo , Proteína p53 Supresora de Tumor/fisiología , Animales , Secuencia de Bases , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Maleatos/farmacología , Datos de Secuencia Molecular , Proteína Quinasa C/metabolismo
10.
Oncogene ; 9(11): 3249-57, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7936649

RESUMEN

Wild-type p53 functions in the G1 DNA damage checkpoint pathway by activating gene transcription and preventing cell cycle progression. Others reported that mutation of the serine 386 codon in mouse p53 abolished its ability to suppress growth. Serine 386 of murine p53 and the homologous residue of human p53, serine 392, are phosphorylated in vivo and can be phosphorylated in vitro by casein kinase II (CKII). We constructed mutants that changed serine 392 of human p53 to alanine (p53-S392A) or aspartic acid (p53-S392D); cotransfection of both these mutants with a reporter gene carrying a p53-responsive element into the p53-null Saos-2 cell line activated transcription as well as did wild-type p53. Furthermore, both mutants blocked cell cycle progression after transient transfection in these cells. A stable derivative of the T98G human glioblastoma cell line was established that expressed p53-S392A in response to dexamethasone. Overexpression of this mutant activated transcription of the endogenous waf1 (also called cip1) and mdm2 genes to the same extent as wild-type p53 and also produced growth arrest. Finally, p53-S392A and p53-S392D suppressed foci formation by activated ras and adenovirus E1A oncogenes as efficiently as did wild-type p53. Thus, unlike mutants that altered the serine 15 phosphorylation site, elimination of the serine 392 phosphorylation site had no discernible effect on p53 function. We conclude that neither phosphorylation nor RNA attachment to serine 392 are required for human p53's ability to suppress cell growth or to activate transcription in vivo.


Asunto(s)
Proteínas Nucleares , Proteínas Proto-Oncogénicas , Serina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas E1A de Adenovirus/genética , Animales , Secuencia de Bases , Sitios de Unión , Quinasa de la Caseína II , Ciclo Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , ADN/metabolismo , Genes ras , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Fosforilación , Inhibidores de Proteínas Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Ratas , Ratas Endogámicas F344 , Activación Transcripcional , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
11.
Proc Natl Acad Sci U S A ; 91(21): 10079-83, 1994 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-7937841

RESUMEN

Overexpression of wild-type p53 protein has been shown to induce arrest in the G1 stage of the cell cycle and to transactivate expression of the gene that encodes the 21-kDa Waf1/Cip1 protein, a potent inhibitor of cyclin-dependent kinase activity. p53-dependent G1 arrest is accompanied by decreased expression of the B-myb gene, a relative of the c-myb cellular oncogene. In this study we show that B-myb expression is required for cells to progress from G1 into S phase and that high levels of ectopic B-myb expression uncoupled from cell cycle regulation rescues cells from p53-induced G1 arrest even in the presence of Waf1/Cip1 transactivation and inhibition of cyclin E/Cdk2 kinase activity. Cotransfection experiments with p53 expression plasmids and expression plasmids encoding in-frame deletion mutations in B-myb coding sequences indicate that the DNA-binding domain of the B-Myb protein is required for this activity. These results provide evidence of a bypass of p53-induced Waf1/Cip1-mediated cell cycle regulatory pathways by a member of the myb oncogene family.


Asunto(s)
Proteínas de Ciclo Celular , Ciclo Celular/fisiología , Ciclinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Oligonucleótidos Antisentido/farmacología , Oncogenes , Inhibidores de Proteínas Quinasas , Transactivadores , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Bases , Northern Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/biosíntesis , Fase G1 , Genes p53 , Humanos , Datos de Secuencia Molecular , Osteosarcoma , Plásmidos , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Factores de Transcripción/biosíntesis , Transfección , Células Tumorales Cultivadas
12.
Proc Natl Acad Sci U S A ; 90(13): 5954-8, 1993 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8327466

RESUMEN

The product of the p53 gene suppresses cell growth and plays a critical role in suppressing development of human tumors. p53 protein binds DNA, activates transcription, and can be phosphorylated at N- and C-terminal sites. Previously, wild-type p53 was shown to be hyperphosphorylated compared to mutant p53 during p53-mediated growth arrest in vivo. Here we show that Ser-15 and Ser-9 in the N-terminal transactivation domain of wild-type human p53 are phosphorylated in vivo in cells derived from the human glioblastoma line T98G. In [Ile237]p53 and [Ala143]p53, two natural p53 mutants from human tumors that are defective for activation of transcription, phosphorylation at Ser-15 was reduced and phosphorylation at Ser-392 was increased compared to wild-type p53. No change was observed at Ser-9. [His273]p53, a third mutant, had a phosphorylation state similar to that of wild-type p53. We suggest that phosphorylation of Ser-15 may depend on the ability of p53 to adopt a wild-type conformation and may contribute to p53's ability to block cell growth.


Asunto(s)
Neoplasias/genética , Serina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Mutación , Neoplasias/metabolismo , Fosfopéptidos/análisis , Fosforilación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
13.
Oncogene ; 8(6): 1519-28, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8502477

RESUMEN

Overexpression of wild-type p53 prevents cells from entering the S phase of the cell cycle. The amino-terminal transactivation region of p53 is phosphorylated by several protein kinases, including DNA-PK, a nuclear serine/threonine protein kinase that in vitro requires DNA for activity. DNA-PK was recently shown to phosphorylate serines 15 and 37 of human p53 (Lees-Miller et al., 1992. Mol. Cell. Biol., 12, 5041-5049). To prevent phosphorylation at these sites, mutants were constructed that changed the codons for serine 15 or serine 37 to alanine codons. Expression of p53-Ala-37 in stably transformed T98G cells blocked progression of the cells into S phase as well as did the expression of wild-type p53. In contrast, p53-Ala-15 was partially defective in blocking cell cycle progression. Several cell clones transformed with the mutant p53-Ala-15 gene expressed normal levels of p53 mRNA but accumulated little or no detectable p53 protein. However, by using a transient expression system driven by a strong cytomegalovirus promoter, we showed that the inability of p53-Ala-15 to fully block cell cycle progression was not due to inadequate levels of expression or to a failure of the mutant protein to accumulate in the nucleus. These results suggest that phosphorylation of Ser-15 may affect p53 function.


Asunto(s)
Ciclo Celular/fisiología , Genes p53 , Serina , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales , Secuencia de Bases , Northern Blotting , Línea Celular , Humanos , Immunoblotting , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oligodesoxirribonucleótidos , Péptidos/síntesis química , Péptidos/inmunología , Fosforilación , Reacción en Cadena de la Polimerasa , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Transfección , Proteína p53 Supresora de Tumor/análisis
14.
Oncogene ; 7(11): 2161-7, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1437144

RESUMEN

The nuclear phosphoprotein p53 is an important regulator of cell proliferation in normal cells. Interestingly, the gene encoding p53 has usually undergone mutations in a wide range of tumor types. Recent studies of the p53 gene in Burkitt's lymphomas have demonstrated that mutations are extremely common, and in fact it is rare that both alleles of the p53 gene in these tumors are not inactivated by mutation or deletion. We present here genetic data regarding the status of the p53 gene in the Burkitt lymphoma cell line, Raji. As is typical for this type of tumor, both alleles have undergone point mutations. Further, statistical analysis of available data from a large number of Burkitt's lymphomas indicates an apparent tumor-specific distribution of p53 mutations. The possibility that specific mutations of the p53 gene may be important for different tumor types is discussed.


Asunto(s)
Linfoma de Burkitt/genética , Genes p53 , Mutación , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Humanos , Datos de Secuencia Molecular
15.
G Batteriol Virol Immunol ; 82(1-12): 82-7, 1989.
Artículo en Italiano | MEDLINE | ID: mdl-2562300

RESUMEN

The effects of interferon (IFN gamma) and of IFN alpha/beta on normal T and B lymphocytes and various T and non-T human and murine cell lines have been investigated. IFN gamma, unlike IFN alpha/beta, did not promote the antiviral state in T cells. The lack of antiviral activity was confirmed at the biochemical level by the finding that 2',5' oligoadenylate synthetase activity is not induced in T cells by IFN gamma only.


Asunto(s)
Interferón gamma/farmacología , Linfocitos T/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/metabolismo , Líquido Amniótico/citología , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Interferón Tipo I/farmacología , Macrófagos/efectos de los fármacos , Ratones , Especificidad de la Especie , Linfocitos T/enzimología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , Virus de la Estomatitis Vesicular Indiana/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA