Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 270
Filtrar
1.
Oncogene ; 26(3): 449-55, 2007 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-16832346

RESUMEN

Normal human diploid fibroblasts (HDFs) are refractory to oncogene-mediated transformations in vitro, compared with rodent fibroblasts. As successful oncogene-mediated transformations of normal HDFs have been reported using the human telomerase catalytic subunit, it has been considered that telomerase activity contributes to the species-specific transformability. However, these transformed HDFs are much less malignant compared with those of rodent cells, suggesting the existence of undefined mechanisms that render HDFs resistant to malignant transformation. Here, cDNA microarray analysis identified caveolin-1 as one of the possible cellular factors involved in such mechanisms. The mitogen-activated protein kinases (MAPK) pathway downregulates Caveolin-1 in rodent fibroblasts, transformed by coexpression of the SV40 early region and activated H-Ras. In contrast, the coexpression of these two oncogenes in HDFs failed to reduce the expression level of Caveolin-1. These results strongly suggest the presence of critical differences in events following the phosphorylation of ERK during the activation process of the MAPK signaling pathway between human and rodent cells, as the ERK protein was similarly phosphorylated in both systems. Furthermore, the small interfering RNA-mediated suppression of Caveolin-1 facilitated the oncogene-mediated transformation of normal HDFs, clearly indicating that the differences in the transformability between human and rodent cells are due, at least in part, to the mechanism responsible for the resistance to Ras-induced Caveolin-1 downregulation in HDFs.


Asunto(s)
Caveolina 1/metabolismo , Transformación Celular Neoplásica , Fibroblastos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo , Animales , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Regulación hacia Abajo , Fibroblastos/citología , Regulación de la Expresión Génica , Humanos , Luciferasas/metabolismo , Pulmón/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , ARN Interferente Pequeño/farmacología , Ratas , Transducción de Señal , Transfección , Proteínas ras/genética
2.
J Exp Clin Cancer Res ; 21(2): 191-5, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12148577

RESUMEN

Ornithine decarboxylase (ODC), a critical regulatory enzyme for polyamine biosynthesis, is strictly regulated in human cells. Several studies suggested the importance of elevated enzymatic activity and altered biochemical characteristics of ODC in malignant cells. Because mutation of ODC in primary human hepatocellular carcinoma has been reported, we examined whether the genetic alterations, such as mutations or structural alterations of the gene, also account for the alteration of ODC activity in human colorectal cancer. No mutation or structural alteration in the ODC was detected in any of the colorectal tumors and normal tissues examined. These results suggest that a mutation or structural alteration of the ODC may not be involved in human colorectal carcinogenesis.


Asunto(s)
Neoplasias Colorrectales/enzimología , Mutación , Ornitina Descarboxilasa/genética , Southern Blotting , Estudios de Casos y Controles , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Cartilla de ADN/química , ADN Complementario/genética , Humanos , Mucosa Intestinal/fisiología , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Polimorfismo Conformacional Retorcido-Simple , ARN Mensajero/genética
3.
EMBO Rep ; 2(9): 814-20, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11520860

RESUMEN

Paxillin is a protein containing four LIM domains, and functions in integrin signaling. We report here that two transcripts are generated from the paxillin gene locus in Drosophila; one encodes a protein homolog of the vertebrate Paxillin (DPxn37), and the other a protein with only three LIM domains, partly encoded by its own specific exon (PDLP). At the myotendinous junctions of Drosophila embryos where integrins play important roles, both DPxn37 and PDLP are highly expressed with different patterns; DPxn37 is predominantly concentrated at the center of the junctions, whereas PDLP is highly enriched at neighboring sides of the junction centers, primarily expressed in the mesodermal myotubes. Northern blot analysis revealed that DPxn37 is ubiquitously expressed throughout the life cycle, whereas PDLP expression exhibits a biphasic pattern during development, largely concomitant with muscle generation and remodeling. Our results collectively reveal that a unique system exists in Drosophila for the generation of a novel type of LIM-only protein, highly expressed in the embryonic musculature, largely utilizing the Paxillin LIM domains.


Asunto(s)
Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Músculos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN/metabolismo , ADN Complementario/metabolismo , Bases de Datos como Asunto , Drosophila , Proteínas de Drosophila , Exones , Biblioteca de Genes , Proteínas Fluorescentes Verdes , Integrinas/metabolismo , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Fluorescente , Modelos Genéticos , Datos de Secuencia Molecular , Oligopéptidos/metabolismo , Paxillin , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal
4.
EMBO Rep ; 2(8): 727-35, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11463744

RESUMEN

FRS2 has been identified in mammalian cells as a protein that is tyrosine phosphorylated and binds to Grb2 and Shp2 in response to fibroblast growth factor (FGF) or nerve growth factor (NGF) stimulation. But neither its existence in other vertebrate classes or invertebrates nor its function during embryonic development has been defined. Here we have identified and characterized a Xenopus homolog of FRS2 (xFRS2). xFRS2 is tyrosine phosphorylated in early embryos, and overexpression of an unphosphorylatable form of xFRS2 interferes with FGF-dependent mesoderm formation. The Src family kinase Laloo, which was shown to function in FGF signaling during early Xenopus development, binds to xFRS2 and promotes tyrosine phosphorylation of xFRS2. Moreover, xFRS2 and Laloo are shown to bind to Xenopus FGF receptor 1. These results suggest that xFRS2 plays an important role in FGF signaling in cooperation with Laloo during embryonic development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Desarrollo Embrionario , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Proteínas de Xenopus , Xenopus laevis/embriología , Familia-src Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo/fisiología , Clonación Molecular , Embrión no Mamífero/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hibridación in Situ , Sustancias Macromoleculares , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación , Unión Proteica , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Alineación de Secuencia , Xenopus laevis/metabolismo , Familia-src Quinasas/genética
5.
Genes Cells ; 6(5): 431-40, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11380621

RESUMEN

BACKGROUND: c-Abl kinase is activated in response to a variety of biological stimuli. Crk family adaptor proteins can interact physically with c-Abl and be involved in the activation of c-Abl kinase. RESULTS: We report that the Crk family of adaptor proteins act as trans-acting activators of c-Abl kinase. The interaction of the amino-terminal Src-homology (SH) 3 domain of c-Crk and the proline-rich motifs of c-Abl is an essential step for the phosphorylation of c-Crk by c-Abl, as well as the activation of c-Abl by c-Crk. The activation of c-Abl by c-Crk is negatively regulated by phosphorylation of the tyrosine 221 of c-Crk. Our data suggest that, in the absence of phosphorylation of the tyrosine Y221, the SH2 domain of c-Crk becomes free to bind to target molecules while the carboxyl-terminal SH3 domain of c-Crk binds to the proline-rich region of c-Abl, inducing the activation of c-Abl by c-Crk. CONCLUSION: This study suggests that the Crk family functions as trans-acting activators of c-Abl kinase. The phosphorylation of c-Crk may regulate c-Abl kinase.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Activación Transcripcional , Dominios Homologos src/genética , Animales , Sitios de Unión , Línea Celular Transformada , Embrión de Pollo , Fibroblastos/metabolismo , Inmunohistoquímica , Ratones , Mutación , Proteína Oncogénica v-crk , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-crk , Proteínas Oncogénicas de Retroviridae/genética , Tirosina/metabolismo
6.
J Biol Chem ; 276(7): 4957-63, 2001 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-11067845

RESUMEN

CMS/CD2AP is a cytoplasmic protein critical for the integrity of the kidney glomerular filtration and the T cell function. CMS contains domains and motifs characteristic for protein-protein interactions, and it is involved in the regulation of the actin cytoskeleton. We report here that the individual SH3 domains of CMS bind to phosphotyrosine proteins of approximately 80, 90, and 180 kDa in cell lysates stimulated with epidermal growth factor. The second SH3 domain of CMS bound specifically to a tyrosine-phosphorylated protein of 120 kDa, which we identified as the proto-oncoprotein c-Cbl. The c-Cbl-binding site for CMS mapped to the carboxyl terminus of c-Cbl and is different from the proline-rich region known to bind SH3-containing proteins. CMS binding to c-Cbl was markedly attenuated in a tyrosine phosphorylation-defective c-Cbl mutant indicating that this interaction is dependent on the tyrosine phosphorylation of CMS. It also implies that CMS interacts with c-Cbl in an inducible fashion upon stimulation of a variety of cell-surface receptors. Immunofluorescence analysis revealed that both proteins colocalize at lamellipodia and leading edges of cells, and we propose that the interaction of CMS with c-Cbl offers a mechanism by which c-Cbl associates and regulates the actin cytoskeleton.


Asunto(s)
Proteínas/química , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitina-Proteína Ligasas , Dominios Homologos src , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células COS , Línea Celular , Proteínas del Citoesqueleto , Humanos , Fosforilación , Fosfotirosina/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-cbl , Seudópodos/metabolismo , Transfección
7.
Proc Natl Acad Sci U S A ; 97(12): 6439-44, 2000 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-10829062

RESUMEN

The kinase activity of Abl is known to be regulated by a putative trans-acting inhibitor molecule interacting with the Src homology (SH) 3 domain of Abl. Here we report that the kinase-deficient Src (SrcKD) directly inhibits the tyrosine phosphorylation of Cbl and other cellular proteins by Abl. We found that both the SH2 and SH3 domains of SrcKD are necessary for the suppressor activity toward the Abl kinase phosphorylating Cbl. To suppress the Cbl phosphorylation by Abl, the interaction between the SH3 domain of SrcKD and Cbl is required. This interaction between SrcKD and Cbl is regulated by a closed structure of Cbl. The binding of Abl to the extreme carboxyl-terminal region of Cbl unmasks the binding site of SrcKD to Cbl. This results in a ternary complex that inhibits the Abl-mediated phosphorylation of Cbl by steric hindrance. These results illustrate a mechanism by which the enzymatically inactive Src can exert a biological function in vivo.


Asunto(s)
Proteínas Proto-Oncogénicas c-abl/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitina-Proteína Ligasas , Familia-src Quinasas/fisiología , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-cbl , Dominios Homologos src
8.
Gene ; 251(1): 37-43, 2000 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-10863094

RESUMEN

The human p107 protein shares many structural and functional features with the retinoblastoma gene product and retinoblastoma-related p130 protein. In this study, we have cloned and elucidated the complete intron-exon organization of the gene encoding the p107 protein. The gene contains 22 exons spanning over 100kilobase pairs of genomic DNA. The length of individual exons ranges from 50 to 840base pairs. The arrays of exons in the p107 gene are rather similar among members of the gene family, especially to those of the p130 gene, while the length of introns is extensively diverse. This study will provide a molecular basis for implementing comprehensive screening for p107 mutations using genomic DNAs from human malignancies. We also show a detailed structure of an intragenic deletion of the p107 gene found in a human B-cell lymphoma cell line, KAL-1, which was shown to occur by homologous recombination between the two directly repeated Alu family sequences.


Asunto(s)
Linfoma de Células B/genética , Proteínas Nucleares/genética , Secuencia de Bases , ADN de Neoplasias/química , ADN de Neoplasias/genética , Exones , Eliminación de Gen , Genes/genética , Humanos , Intrones , Linfoma de Células B/patología , Proteína p107 Similar a la del Retinoblastoma , Análisis de Secuencia de ADN , Eliminación de Secuencia , Células Tumorales Cultivadas
9.
Cancer Res ; 60(12): 3143-6, 2000 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10866301

RESUMEN

We characterized the genomic structure of the human ING1 gene, a candidate tumor suppressor gene, and found that the gene has three exons. We also demonstrated that four mRNA variants were transcribed from three different promoter regions. Of 34 informative cases of head and neck squamous cell carcinoma, 68% of tumors showed loss of heterozygosity at chromosome 13q33-34, where the ING1 gene is located. Here we present the first report that three missense mutations and three silent changes were detected in the ING1 gene in 6 of 23 tumors with allelic loss at the 13q33-34 region. These missense mutations were found within the PHD finger domain and nuclear localization motif in ING1 protein, probably abrogating the normal function.


Asunto(s)
Carcinoma de Células Escamosas/genética , Genes Supresores de Tumor , Neoplasias de Cabeza y Cuello/genética , Mutación Missense , Proteínas/genética , Proteínas de Ciclo Celular , Cromosomas Humanos Par 13 , Proteínas de Unión al ADN , Exones , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular , Pérdida de Heterocigocidad , Luciferasas/metabolismo , Repeticiones de Microsatélite , Modelos Genéticos , Proteínas Nucleares , Polimorfismo Conformacional Retorcido-Simple , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor
10.
Proc Natl Acad Sci U S A ; 97(13): 7290-5, 2000 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-10852971

RESUMEN

v-Crk induces cellular tyrosine phosphorylation and transformation of chicken embryo fibroblasts (CEF). We studied the molecular mechanism of the v-Crk-induced transformation. Experiments with Src homology (SH)2 and SH3 domain mutants revealed that the induction of tyrosine phosphorylation of cellular proteins requires only the SH2 domain, but both the SH2 and SH3 domains are required for complete transformation. Analysis of three well defined signaling pathways, the mitogen-activated protein kinase (MAPK) pathway, the Jun N-terminal kinase (JNK) pathway, and the phosphoinositide 3-kinase (PI3K)/AKT pathway, demonstrated that only the PI3K/AKT pathway is constitutively activated in v-Crk-transformed CEF. Both the SH2 and SH3 domains are required for this activation of the PI3K/AKT pathway in CEF. We also found that the colony formation of CEF is strongly induced by a constitutively active PI3K mutant, and that a PI3K inhibitor, LY294002, suppresses the v-Crk-induced transformation. These results strongly suggest that constitutive activation of the PI3K/AKT pathway plays an essential role in v-Crk-induced transformation of CEF.


Asunto(s)
Transformación Celular Neoplásica , Fibroblastos/patología , Fibroblastos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Oncogénicas de Retroviridae/fisiología , Transducción de Señal , Animales , Células Cultivadas , Pollos , Datos de Secuencia Molecular , Proteína Oncogénica v-crk
11.
Cancer Res ; 60(9): 2361-4, 2000 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10811109

RESUMEN

To search for the intracellular signaling pathway critical for the secretion of matrix metalloproteinases (MMP), we studied the effects of dominant negative Ras (S17N Ras) and dominant negative MEK1 (MEK1AA) expression in v-crk-transformed 3Y1. Expression of either S17N Ras or MEK1AA dramatically suppressed the augmented secretion of MMP-2 and MMP-9 in v-crk-transfected 3Y1. Similarly, a Ras farnesyltransferase inhibitor, manumycin A, and a MEK1 inhibitor, U0126, suppressed MMP secretion in a dose-dependent manner, whereas a PI3 kinase inhibitor, wortmannin, could not. In addition, the suppression of MMP secretion by S17N Ras showed good correlation with the inhibition of in vitro invasiveness of the cells. In contrast, expression of dominant negative C3G did not suppress MMP secretion, although it substantially blocked the c-Jun N-terminal kinase activation. Taken together, the Ras-MEK1 pathway, but not the C3G-JNK pathway, seems to play a key role in the activation of MMP secretion and, hence, the invasiveness of v-crk-transformed cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Metaloproteinasas de la Matriz/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Proteínas ras/metabolismo , Androstadienos/farmacología , Animales , Butadienos/farmacología , Línea Celular Transformada , Colágeno/metabolismo , Combinación de Medicamentos , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Fibroblastos/enzimología , Factor 2 Liberador de Guanina Nucleótido/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Immunoblotting , Laminina/metabolismo , MAP Quinasa Quinasa 1 , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Proteína Oncogénica v-crk , Polienos/farmacología , Alcamidas Poliinsaturadas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoglicanos/metabolismo , Ratas , Transducción de Señal , Wortmanina , Dominios Homologos src
12.
EMBO Rep ; 1(1): 32-9, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11256621

RESUMEN

Although a number of genes that are involved in the establishment of left-right asymmetry have been identified, earlier events in the molecular pathway developing left-right asymmetry remain to be elucidated. Here we present evidence suggesting that the transforming growth factor-beta family member derrière is involved in the development of left-right asymmetry in Xenopus embryos. Ectopic expression of derrière on the right side can fully invert cardiac and visceral left-right orientation and nodal expression, and expression of a dominant-negative form of derrière on the left side can partially randomize the left-right orientation and nodal expression. Moreover, while expression of the dominant-negative derrière does not inhibit the activity of Vg1 directly, it can rescue the altered left-right orientation induced by Vg1. Vg1 can induce derrière in animal cap explants. These results suggest that derrière is involved in earlier molecular pathways developing the left-right asymmetry.


Asunto(s)
Tipificación del Cuerpo/genética , Desarrollo Embrionario , Sustancias de Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus , Animales , Western Blotting , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/fisiología , Femenino , Glicoproteínas/genética , Glicoproteínas/metabolismo , Sustancias de Crecimiento/genética , Corazón/embriología , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Microinyecciones , Datos de Secuencia Molecular , Oocitos/fisiología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/genética , Vísceras/embriología , Xenopus , Proteínas de Pez Cebra
13.
Cancer Res ; 60(24): 7033-8, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11156408

RESUMEN

PTEN is a tumor suppressor frequently inactivated in brain, prostate, and uterine cancer. It acts as a phosphoinositide phosphatase and consists of an amino-terminal phosphatase domain tightly linked to a COOH-terminal C2 domain involved in lipid membrane-binding. We investigated the functions of the C2 domain and their relevance for tumor growth. To discriminate between PTEN C2 domain ability to recruit or to position the active site to the membrane, we artificially membrane-targeted PTEN by a myristoylation signal. This modification increased wild-type PTEN growth inhibition but did not rescue a C2 mutant defective in lipid-binding, suggesting a model in which PTEN C2 domain positions the active site productively with respect to the membrane-bound phosphoinositide substrate. When tumor-derived mutations in the loops that connect the C2 beta-strands were analyzed, we found that these generally destabilized the protein but had variable effects on the phosphatase activity and tumor growth. The magnitude of these effects was dependent on the presence of the COOH-terminal PEST sequences and on the cell type where the mutant proteins were expressed, suggesting the existence of fluctuating structural defects of the mutant protein. One of the C2 loop mutants induced a total loss of PTEN tumor-suppressor function, most likely by affecting both the membrane binding and the protein stability. These data support a double role for PTEN C2 domain in protein stability and in productive orientation of the catalytic site.


Asunto(s)
Monoéster Fosfórico Hidrolasas/química , Proteínas Supresoras de Tumor , Sitios de Unión , Dominio Catalítico , División Celular , Membrana Celular/metabolismo , Eliminación de Gen , Humanos , Immunoblotting , Metabolismo de los Lípidos , Microscopía Fluorescente , Modelos Moleculares , Mutación , Ácidos Mirísticos/metabolismo , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/metabolismo , Plásmidos/metabolismo , Mutación Puntual , Pruebas de Precipitina , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transfección , Células Tumorales Cultivadas
14.
Proc Natl Acad Sci U S A ; 96(18): 10182-7, 1999 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-10468583

RESUMEN

PTEN is a recently identified tumor suppressor inactivated in a variety of cancers such as glioblastoma and endometrial and prostate carcinoma. It contains an amino-terminal phosphatase domain and acts as a phosphatidylinositol 3,4,5-trisphosphate phosphatase antagonizing the activity of the phosphatidylinositol 3-OH kinase. PTEN also contains a carboxyl-terminal domain, and we addressed the role of this region that, analogous to the amino-terminal phosphatase domain, is the target of many mutations identified in tumors. Expression of carboxyl-terminal mutants in PTEN-deficient glioblastoma cells permitted the anchorage-independent growth of the cells that otherwise was suppressed by wild-type PTEN. The stability of these mutants in cells was reduced because of rapid degradation. Although the carboxyl-terminal region contains regulatory PEST sequences and a PDZ-binding motif, these specific elements were dispensable for the tumor-suppressor function. The study of carboxyl-terminal point mutations affecting the stability of PTEN revealed that these were located in strongly predicted beta-strands. Surprisingly, the phosphatase activity of these mutants was affected in correlation with the degree of disruption of these structural elements. We conclude that the carboxyl-terminal region is essential for regulating PTEN stability and enzymatic activity and that mutations in this region are responsible for the reversion of the tumor-suppressor phenotype. We also propose that the molecular conformational changes induced by these mutations constitute the mechanism for PTEN inactivation.


Asunto(s)
Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Supresoras de Tumor , Secuencia de Aminoácidos , Animales , Células COS , Femenino , Genes Supresores de Tumor , Glioblastoma , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/genética , Placenta/metabolismo , Mutación Puntual , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfección , Células Tumorales Cultivadas
15.
J Biol Chem ; 274(38): 27161-7, 1999 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-10480932

RESUMEN

Transforming growth factor-beta (TGF-beta)-activated kinase 1 (TAK1), a member of the mitogen-activated protein kinase kinase kinase family, is suggested to be involved in TGF-beta-induced gene expression, but the signaling mechanism from TAK1 to the nucleus remains largely undefined. We have found that p38 mitogen-activated protein kinase, and its direct activator MKK6 are rapidly activated in response to TGF-beta. Expression of dominant negative MKK6 or dominant negative TAK1 inhibited the TGF-beta-induced transcriptional activation as well as the p38 activation. Constitutive activation of the p38 pathway in the absence of TGF-beta induced the transcriptional activation, which was enhanced synergistically by coexpression of Smad2 and Smad4 and was inhibited by expression of the C-terminal truncated, dominant negative Smad4. Furthermore, we have found that activating transcription factor-2 (ATF-2), which is known as a nuclear target of p38, becomes phosphorylated in the N-terminal activation domain in response to TGF-beta, that ATF-2 forms a complex with Smad4, and that the complex formation is enhanced by TGF-beta. In addition, expression of a nonphosphorylatable form of ATF-2 inhibited the TGF-beta-induced transcriptional activation. These results show that the p38 pathway is activated by TGF-beta and is involved in the TGF-beta-induced transcriptional activation by regulating the Smad-mediated pathway.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Regulación de la Expresión Génica , Quinasas Quinasa Quinasa PAM , Proteínas Quinasas Activadas por Mitógenos , Factor de Crecimiento Transformador beta/fisiología , Factor de Transcripción Activador 2 , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes Supresores de Tumor , Leucina Zippers , MAP Quinasa Quinasa 6 , Proteínas Quinasas/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
16.
Proc Natl Acad Sci U S A ; 96(11): 6211-6, 1999 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-10339567

RESUMEN

Cas ligand with multiple Src homology (SH) 3 domains (CMS) is an ubiquitously expressed signal transduction molecule that interacts with the focal adhesion protein p130(Cas). CMS contains three SH3 in its NH2 terminus and proline-rich sequences in its center region. The latter sequences mediate the binding to the SH3 domains of p130(Cas), Src-family kinases, p85 subunit of phosphatidylinositol 3-kinase, and Grb2. The COOH-terminal region contains putative actin binding sites and a coiled-coil domain that mediates homodimerization of CMS. CMS is a cytoplasmic protein that colocalizes with F-actin and p130(Cas) to membrane ruffles and leading edges of cells. Ectopic expression of CMS in COS-7 cells resulted in alteration in arrangement of the actin cytoskeleton. We observed a diffuse distribution of actin in small dots and less actin fiber formation. Altogether, these features suggest that CMS functions as a scaffolding molecule with a specialized role in regulation of the actin cytoskeleton.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto , Citoesqueleto/fisiología , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas , Transcripción Genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Secuencia de Aminoácidos , Animales , Células COS , Proteínas Portadoras/química , Línea Celular , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Proteína Sustrato Asociada a CrK , Citoesqueleto/ultraestructura , Femenino , Feto , Biblioteca de Genes , Humanos , Datos de Secuencia Molecular , Especificidad de Órganos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Transfección
17.
J Biol Chem ; 274(17): 12163-70, 1999 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-10207044

RESUMEN

Smad family proteins have been identified as mediators of intracellular signal transduction by the transforming growth factor-beta (TGF-beta) superfamily. Each member of the pathway-restricted, receptor-activated Smad family cooperates and synergizes with Smad4, called co-Smad, to transduce the signals. Only Smad4 has been shown able to function as a common partner of the various pathway-restricted Smads in mammals. Here we have identified a novel Smad4-like molecule in Xenopus (XSmad4beta) as well as a Xenopus homolog of a well established Smad4 (XSmad4alpha). XSmad4beta is 70% identical to XSmad4alpha in amino acid sequence. Both of the Xenopus Smad4s can cooperate with Smad1 and Smad2, the pathway-restricted Smads specific for bone morphogenetic protein and TGF-beta, respectively. However, they show distinct properties in terms of their developmental expression patterns, subcellular localizations, and phosphorylation states. Moreover, XSmad4beta, but not XSmad4alpha, has the potent ability to induce ventralization when microinjected into the dorsal marginal region of the 4-cell stage of the embryos. These results suggest that the two Xenopus Smad4s have overlapping but distinct functions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Isoformas de Proteínas/metabolismo , Transactivadores/metabolismo , Proteínas de Xenopus , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN Complementario , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Homología de Secuencia de Aminoácido , Proteínas Smad , Proteína Smad4 , Fracciones Subcelulares/metabolismo , Transactivadores/química , Transactivadores/genética , Xenopus
18.
Mol Cell Neurosci ; 13(1): 9-23, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10049528

RESUMEN

Semaphorins comprise a large family of proteins implicated in axonal guidance. We cloned a novel transmembrane semaphorin, semaphorin Y (Sema Y), which has a class VI sema domain. Sema Y shows growth cone collapsing activity on DRG neurons in vitro, and the target regions of the DRG neurons express sema Y mRNA during development. Sema Y may be a stop signal for these neurons in their target areas. Interestingly, sema Y mRNA was also detected in other neurons and their targets. Two isoforms of Sema Y derived from alternative splicing were identified and their expression was found to be regulated in a tissue- and age-dependent manner. Distribution of sema Y mRNA suggests that Sema Y might also be important during maintenance of axonal connections and/or differentiation and migration of cells. Sequence comparison among class VI semaphorins revealed two short conserved sequence stretches in their cytoplasmic domains, suggesting interaction of these semaphorins with a common intracellular component(s).


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Clonación Molecular , Empalme Alternativo , Secuencia de Aminoácidos/genética , Animales , Moléculas de Adhesión Celular Neuronal/fisiología , Glicoproteínas/fisiología , Conos de Crecimiento/fisiología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Semaforinas
20.
EMBO J ; 18(1): 145-55, 1999 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-9878058

RESUMEN

The cellular signal transduction pathways by which C3G, a RAS family guanine nucleotide exchange factor, mediates v-crk transformation are not well understood. Here we report the identification of Drosophila C3G, which, like its human cognate, specifically binds to CRK but not DRK/GRB2 adaptor molecules. During Drosophila development, constitutive membrane binding of C3G, which also occurs during v-crk transformation, results in cell fate changes and overproliferation, mimicking overactivity of the RAS-MAPK pathway. The effects of C3G overactivity can be suppressed by reducing the gene dose of components of the RAS-MAPK pathway and of RAP1. These findings provide the first in vivo evidence that membrane localization of C3G can trigger activation of RAP1 and RAS resulting in the activation of MAPK, one of the hallmarks of v-crk transformation previously thought to be mediated through activation of SOS.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Insectos/metabolismo , Proteínas/metabolismo , Proteínas ras/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , División Celular , Mapeo Cromosómico , Cartilla de ADN/genética , Drosophila/citología , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido , Humanos , Proteínas de Insectos/genética , Datos de Secuencia Molecular , Proteína Oncogénica v-crk , Proteínas/genética , Proteínas Oncogénicas de Retroviridae/metabolismo , Transducción de Señal , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo , Proteínas de Unión al GTP rap , Factores de Intercambio de Guanina Nucleótido ras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...