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.
Artigo em Inglês | MEDLINE | ID: mdl-16832346

RESUMO

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.


Assuntos
Caveolina 1/metabolismo , Transformação Celular Neoplásica , Fibroblastos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas ras/metabolismo , Animais , Caveolina 1/antagonistas & inibidores , Caveolina 1/genética , Regulação para Baixo , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Luciferases/metabolismo , Pulmão/metabolismo , Camundongos , Células NIH 3T3 , Fosforilação , RNA Interferente Pequeno/farmacologia , Ratos , Transdução de Sinais , Transfecção , Proteínas ras/genética
2.
J Exp Clin Cancer Res ; 21(2): 191-5, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12148577

RESUMO

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.


Assuntos
Neoplasias Colorretais/enzimologia , Mutação , Ornitina Descarboxilase/genética , Southern Blotting , Estudos de Casos e Controles , Neoplasias Colorretais/genética , Análise Mutacional de DNA , Primers do DNA/química , DNA Complementar/genética , Humanos , Mucosa Intestinal/fisiologia , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Polimorfismo Conformacional de Fita Simples , RNA Mensageiro/genética
3.
EMBO Rep ; 2(9): 814-20, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11520860

RESUMO

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.


Assuntos
Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Músculos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA/metabolismo , DNA Complementar/metabolismo , Bases de Dados como Assunto , Drosophila , Proteínas de Drosophila , Éxons , Biblioteca Gênica , Proteínas de Fluorescência Verde , Integrinas/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , Oligopeptídeos/metabolismo , Paxilina , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais
4.
EMBO Rep ; 2(8): 727-35, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11463744

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Desenvolvimento Embrionário , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Xenopus , Xenopus laevis/embriologia , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Padronização Corporal/fisiologia , Clonagem Molecular , Embrião não Mamífero/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hibridização In Situ , Substâncias Macromoleculares , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , Ligação Proteica , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Alinhamento de Sequência , Xenopus laevis/metabolismo , Quinases da Família src/genética
5.
Genes Cells ; 6(5): 431-40, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11380621

RESUMO

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.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Ativação Transcricional , Domínios de Homologia de src/genética , Animais , Sítios de Ligação , Linhagem Celular Transformada , Embrião de Galinha , Fibroblastos/metabolismo , Imuno-Histoquímica , Camundongos , Mutação , Proteína Oncogênica v-crk , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-crk , Proteínas Oncogênicas de Retroviridae/genética , Tirosina/metabolismo
6.
J Biol Chem ; 276(7): 4957-63, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11067845

RESUMO

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.


Assuntos
Proteínas/química , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases , Domínios de Homologia de src , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células COS , Linhagem Celular , Proteínas do Citoesqueleto , Humanos , Fosforilação , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas c-cbl , Pseudópodes/metabolismo , Transfecção
7.
Gene ; 251(1): 37-43, 2000 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-10863094

RESUMO

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.


Assuntos
Linfoma de Células B/genética , Proteínas Nucleares/genética , Sequência de Bases , DNA de Neoplasias/química , DNA de Neoplasias/genética , Éxons , Deleção de Genes , Genes/genética , Humanos , Íntrons , Linfoma de Células B/patologia , Proteína p107 Retinoblastoma-Like , Análise de Sequência de DNA , Deleção de Sequência , Células Tumorais Cultivadas
8.
Proc Natl Acad Sci U S A ; 97(13): 7290-5, 2000 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-10852971

RESUMO

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.


Assuntos
Transformação Celular Neoplásica , Fibroblastos/patologia , Fibroblastos/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Transdução de Sinais , Animais , Células Cultivadas , Galinhas , Dados de Sequência Molecular , Proteína Oncogênica v-crk
9.
Cancer Res ; 60(12): 3143-6, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10866301

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Genes Supressores de Tumor , Neoplasias de Cabeça e Pescoço/genética , Mutação de Sentido Incorreto , Proteínas/genética , Proteínas de Ciclo Celular , Cromossomos Humanos Par 13 , Proteínas de Ligação a DNA , Éxons , Humanos , Proteína 1 Inibidora do Crescimento , Peptídeos e Proteínas de Sinalização Intracelular , Perda de Heterozigosidade , Luciferases/metabolismo , Repetições de Microssatélites , Modelos Genéticos , Proteínas Nucleares , Polimorfismo Conformacional de Fita Simples , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor
10.
Proc Natl Acad Sci U S A ; 97(12): 6439-44, 2000 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-10829062

RESUMO

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.


Assuntos
Proteínas Proto-Oncogênicas c-abl/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases , Quinases da Família src/fisiologia , Sequência de Aminoácidos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas c-cbl , Domínios de Homologia de src
11.
Cancer Res ; 60(9): 2361-4, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811109

RESUMO

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.


Assuntos
Sistema de Sinalização das MAP Quinases , Metaloproteinases da Matriz/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Proteínas ras/metabolismo , Androstadienos/farmacologia , Animais , Butadienos/farmacologia , Linhagem Celular Transformada , Colágeno/metabolismo , Combinação de Medicamentos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/enzimologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Immunoblotting , Laminina/metabolismo , MAP Quinase Quinase 1 , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Proteína Oncogênica v-crk , Polienos/farmacologia , Alcamidas Poli-Insaturadas , Proteínas Serina-Treonina Quinases/metabolismo , Proteoglicanas/metabolismo , Ratos , Transdução de Sinais , Wortmanina , Domínios de Homologia de src
12.
EMBO Rep ; 1(1): 32-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11256621

RESUMO

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.


Assuntos
Padronização Corporal/genética , Desenvolvimento Embrionário , Substâncias de Crescimento/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Xenopus , Animais , Western Blotting , Embrião não Mamífero/anatomia & histologia , Embrião não Mamífero/fisiologia , Feminino , Glicoproteínas/genética , Glicoproteínas/metabolismo , Substâncias de Crescimento/genética , Coração/embriologia , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular , Microinjeções , Dados de Sequência Molecular , Oócitos/fisiologia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/genética , Vísceras/embriologia , Xenopus , Proteínas de Peixe-Zebra
13.
Cancer Res ; 60(24): 7033-8, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11156408

RESUMO

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.


Assuntos
Monoéster Fosfórico Hidrolases/química , Proteínas Supressoras de Tumor , Sítios de Ligação , Domínio Catalítico , Divisão Celular , Membrana Celular/metabolismo , Deleção de Genes , Humanos , Immunoblotting , Metabolismo dos Lipídeos , Microscopia de Fluorescência , Modelos Moleculares , Mutação , Ácidos Mirísticos/metabolismo , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/metabolismo , Plasmídeos/metabolismo , Mutação Puntual , Testes de Precipitina , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transfecção , Células Tumorais Cultivadas
14.
Proc Natl Acad Sci U S A ; 96(18): 10182-7, 1999 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10468583

RESUMO

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.


Assuntos
Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Animais , Células COS , Feminino , Genes Supressores de Tumor , Glioblastoma , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Placenta/metabolismo , Mutação Puntual , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfecção , Células Tumorais Cultivadas
15.
J Biol Chem ; 274(38): 27161-7, 1999 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-10480932

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Regulação da Expressão Gênica , MAP Quinase Quinase Quinases , Proteínas Quinases Ativadas por Mitógeno , Fator de Crescimento Transformador beta/fisiologia , Fator 2 Ativador da Transcrição , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genes Supressores de Tumor , Zíper de Leucina , MAP Quinase Quinase 6 , Proteínas Quinases/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Proc Natl Acad Sci U S A ; 96(11): 6211-6, 1999 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10339567

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto , Citoesqueleto/fisiologia , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/química , Linhagem Celular , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Proteína Substrato Associada a Crk , Citoesqueleto/ultraestrutura , Feminino , Feto , Biblioteca Gênica , Humanos , Dados de Sequência Molecular , Especificidade de Órgãos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína p130 Retinoblastoma-Like , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Transfecção
17.
J Biol Chem ; 274(17): 12163-70, 1999 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-10207044

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Isoformas de Proteínas/metabolismo , Transativadores/metabolismo , Proteínas de Xenopus , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Dados de Sequência Molecular , Fatores de Crescimento Neural , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Homologia de Sequência de Aminoácidos , Proteínas Smad , Proteína Smad4 , Frações Subcelulares/metabolismo , Transativadores/química , Transativadores/genética , Xenopus
18.
Mol Cell Neurosci ; 13(1): 9-23, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10049528

RESUMO

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).


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Clonagem Molecular , Processamento Alternativo , Sequência de Aminoácidos/genética , Animais , Moléculas de Adesão Celular Neuronais/fisiologia , Glicoproteínas/fisiologia , Cones de Crescimento/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Semaforinas
20.
EMBO J ; 18(1): 145-55, 1999 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-9878058

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Insetos/metabolismo , Proteínas/metabolismo , Proteínas ras/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Divisão Celular , Mapeamento Cromossômico , Primers do DNA/genética , Drosophila/citologia , Olho/crescimento & desenvolvimento , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Troca do Nucleotídeo Guanina , Humanos , Proteínas de Insetos/genética , Dados de Sequência Molecular , Proteína Oncogênica v-crk , Proteínas/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , Transdução de Sinais , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo , Proteínas rap de Ligação ao GTP , Fatores ras de Troca de Nucleotídeo Guanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA