Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biol ; 148(6): 1151-8, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10725328

RESUMO

Integrin-mediated leukocyte adhesion is a critical aspect of leukocyte function that is tightly regulated by diverse stimuli, including chemokines, antigen receptors, and adhesion receptors. How cellular signals from CD31 and other adhesion amplifiers are integrated with those from classical mitogenic stimuli to regulate leukocyte function remains poorly understood. Here, we show that the cytoplasmic tail of CD31, an important integrin adhesion amplifier, propagates signals that induce T cell adhesion via beta1 (VLA-4) and beta2 (LFA-1) integrins. We identify the small GTPase, Rap1, as a critical mediator of this effect. Importantly, CD31 selectively activated the small Ras-related GTPase, Rap1, but not Ras, R-Ras, or Rap2. An activated Rap1 mutant stimulated T lymphocyte adhesion to intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM), as did the Rap1 guanine nucleotide exchange factor C3G and a catalytically inactive mutant of RapGAP. Conversely, negative regulators of Rap1 signaling blocked CD31-dependent adhesion. These findings identify a novel important role for Rap1 in regulating ligand-induced cell adhesion and suggest that Rap1 may play a more general role in coordinating adhesion-dependent signals during leukocyte migration and extravasation. Our findings also suggest an alternative mechanism, distinct from interference with Ras-proximal signaling, by which Rap1 might mediate transformation reversion.


Assuntos
Moléculas de Adesão Celular/metabolismo , Adesão Celular/fisiologia , Integrinas/fisiologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Receptores de Retorno de Linfócitos/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Antígenos CD/fisiologia , Humanos , Integrina alfa4beta1 , Células Jurkat , Molécula-1 de Adesão Celular Endotelial a Plaquetas/química , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Linfócitos T/fisiologia , Transfecção , Molécula 1 de Adesão de Célula Vascular/metabolismo
2.
Trends Biochem Sci ; 20(1): 18-22, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7878738

RESUMO

A powerful combination of genetics and biochemistry has provided details of how Ras-directed signalling interacts with and is regulated by other cellular signalling pathways. This might ultimately lead to the control of deregulated signalling by oncogenic Ras. Recently, progress has been made in understanding the regulation of Ras-mediated activation of the Raf-1-ERK2 kinase cascade through crosstalk with protein kinase C and cyclic-AMP-dependent protein kinase.


Assuntos
Genes ras , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/genética , Ativação Enzimática , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-raf
3.
Curr Opin Genet Dev ; 9(1): 112-7, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10072355

RESUMO

The Ral guanine nucleotide exchange factors are direct targets of Ras, providing a mechanism for Ral activation by extracellular signals. In addition, Ral can be activated by a Ras-independent pathway. Ral guanine nucleotide exchange factors contribute to cellular transformation induced by oncogenic Ras through an Erk-independent mechanism which may involve activation of transcription.


Assuntos
Proteínas ras/fisiologia , Ativação Enzimática , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Guanosina Trifosfato/metabolismo , Humanos , Proteínas/metabolismo , Proteínas ral de Ligação ao GTP , Proteínas rap de Ligação ao GTP , Fatores ras de Troca de Nucleotídeo Guanina
4.
Mol Biol Cell ; 16(1): 106-16, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15525675

RESUMO

The Rap-pathway has been implicated in various cellular processes but its exact physiological function remains poorly defined. Here we show that the Caenorhabditis elegans homologue of the mammalian guanine nucleotide exchange factors PDZ-GEFs, PXF-1, specifically activates Rap1 and Rap2. Green fluorescent protein (GFP) reporter constructs demonstrate that sites of pxf-1 expression include the hypodermis and gut. Particularly striking is the oscillating expression of pxf-1 in the pharynx during the four larval molts. Deletion of the catalytic domain from pxf-1 leads to hypodermal defects, resulting in lethality. The cuticle secreted by pxf-1 mutants is disorganized and can often not be shed during molting. At later stages, hypodermal degeneration is seen and animals that reach adulthood frequently die with a burst vulva phenotype. Importantly, disruption of rap-1 leads to a similar, but less severe phenotype, which is enhanced by the simultaneous removal of rap-2. In addition, the lethal phenotype of pxf-1 can be rescued by expression of an activated version of rap-1. Together these results demonstrate that the pxf-1/rap pathway in C. elegans is required for maintenance of epithelial integrity, in which it probably functions in polarized secretion.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Epitélio/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Domínio Catalítico , Proliferação de Células , DNA Complementar/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Genes Reporter , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Homozigoto , Microscopia Eletrônica , Microscopia de Fluorescência , Modelos Genéticos , Mutação , Fenótipo
5.
Oncogene ; 25(5): 657-64, 2006 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-16170341

RESUMO

The mTOR/S6K/4E-BP1 pathway integrates extracellular signals derived from growth factors, and intracellular signals, determined by the availability of nutrients like amino acids and glucose. Activation of this pathway requires inhibition of the tumor suppressor complex TSC1/2. TSC2 is a GTPase-activating protein for the small GTPase Ras homologue enriched in brain (Rheb), GTP loading of which activates mTOR by a yet unidentified mechanism. The level at which this pathway senses the availability of amino acids is unknown but is suggested to be at the level of TSC2. Here, we show that amino-acid depletion completely blocks insulin- and TPA-induced Rheb activation. This indicates that amino-acid sensing occurs upstream of Rheb. Despite this, amino-acid depletion can still inhibit mTOR/S6 kinase signaling in TSC2-/- fibroblasts. Since under these conditions Rheb-GTP levels remain high, a second level of amino-acid sensing exists, affecting mTOR activity in a Rheb-independent fashion.


Assuntos
Aminoácidos/fisiologia , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Neuropeptídeos/fisiologia , Linhagem Celular , Fosforilação , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
6.
Curr Biol ; 8(8): 471-4, 1998 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-9550702

RESUMO

The small GTPase Ral is a Ras-like GTPase [1] that has been implicated in growth-factor-induced and Ras-induced DNA synthesis [2-4], and Ras-induced oncogenic transformation [3,5]. Recently, we and others found that three different Ral guanine nucleotide exchange factors (Ral GEFs) - Ral GDS, Rgl and Rlf - bind specifically to the GTP-bound form of several Ras-like GTPases [6-9]. Although oncogenic Ras is able to activate these Ral GEFs [2,5,10], it is unknown whether growth factors can induce the activation of Ral and, if so, which small GTPase is involved in this process. Here, we show that stimulation of various growth factor receptors, including receptor tyrosine kinases and serpentine receptors, results in rapid activation of Ral. This activation correlates with the activation of Ras, and dominant-negative Ras completely inhibits Ral activation induced by insulin and epidermal growth factor (EGF). From these results, we conclude that Ral activation is a direct downstream effect of growth-factor-induced Ras activation.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Genes ras/fisiologia , Células 3T3 , Animais , Linhagem Celular , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos , Insulina/farmacologia , Lisofosfolipídeos/farmacologia , Camundongos , Mutação , Ratos , Receptores de Endotelina/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão , Proteínas ral de Ligação ao GTP
7.
Mol Cell Biol ; 11(12): 5963-7, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1658621

RESUMO

Insulin induces a rapid activation of p21ras in NIH 3T3 and Chinese hamster ovary cells that overexpress the insulin receptor. Previously, we suggested that p21ras may mediate insulin-induced gene expression. To test such a function of p21ras more directly, we studied the effect of different dominant inhibitory mutants of p21ras on the induction of gene expression in response to insulin. We transfected a collagenase promoter-chloramphenicol acetyltransferase (CAT) gene or a fos promoter-luciferase gene into NIH 3T3 cells that overexpressed the insulin receptor. The activities of both promoters were strongly induced after treatment with insulin. This induction could be suppressed by cotransfection of two inhibitory mutant ras genes, H-ras(Asn-17) or H-ras(Leu-61,Ser-186). In particular, insulin-induced activation of the fos promoter was inhibited completely by H-ras(Asn-17). These results show that p21ras functions as an intermediate in the insulin signal transduction route leading to the induction of gene expression.


Assuntos
Regulação da Expressão Gênica , Insulina/fisiologia , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Células 3T3 , Animais , Linhagem Celular , Cloranfenicol O-Acetiltransferase/genética , Clonagem Molecular , Genes Dominantes , Células HeLa , Humanos , Camundongos , Colagenase Microbiana/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor de Insulina/biossíntese , Receptor de Insulina/genética , Transdução de Sinais , Transfecção
8.
Mol Cell Biol ; 14(11): 7078-85, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7935423

RESUMO

Human primary keratinocytes are an elegant model system to study the balance between proliferation and differentiation. Both epidermal growth factor (EGF) and extracellular calcium have been implicated to function in the control of this balance, although the molecular mechanism underlying this process is poorly understood. In this study, we measured the effect of both EGF and calcium treatment on activation of p21ras and ERK2. We found that addition of EGF stimulated the activity of ERK2. This stimulation was dependent on p21ras activity, since it was completely abolished by expression of a dominant negative mutant of p21ras (p21ras(Asn-17)). Raising the level of extracellular calcium (1.8 mM) did not result in activation of ERK2. On the contrary, calcium treatment inhibited EGF-induced stimulation of ERK2 activity. In order to determine the site at which calcium treatment interferes in EGF-induced signaling, we analyzed the effect of calcium on the various steps that are involved in EGF-induced, p21ras-dependent activation of ERK2. We observed that calcium treatment inhibited EGF-induced p21ras activation. Calcium treatment, however, did not interfere with EGF-induced EGF receptor autophosphorylation or association of mammalian SOS with the EGF receptor and Shc. This, together with the observation that calcium treatment alone decreased the basal level of p21ras activity, indicates that calcium treatment interferes in EGF-mediated signaling at the level of p21ras. This type of cross talk may play a role in the decision between proliferation and differentiation in human primary keratinocytes.


Assuntos
Cálcio/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular , Divisão Celular , Células Cultivadas , Receptores ErbB/metabolismo , Proteínas Ativadoras de GTPase , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno , Modelos Biológicos , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Ativadoras de ras GTPase
9.
Mol Cell Biol ; 19(5): 3885-94, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10207112

RESUMO

Multiple adenovirus (Ad) early proteins have been shown to inhibit transcription activation by p53 and thereby to alter its normal biological functioning. Since these Ad proteins affect the activity of p53 via different mechanisms, we examined whether this inhibition is target gene specific. In addition, we analyzed whether the same Ad early proteins have a comparable effect on transcription activation by the recently identified p53 homologue p73. Our results show that the large E1B proteins very efficiently inhibited the activity of p53 on the Bax, p21(Waf1), cyclin G, and MDM2 reporter constructs but had no effect on the activation of the same reporter constructs by p73, with the exception of some inhibition of the Bax promoter by Ad12 E1B. The repressive effect of the E1A proteins on p53 activity is less than that seen with the large E1B proteins, but the E1A proteins inhibit the activity of both p53 and p73. We could not detect significant inhibition of p53 functions by E4orf6, but a clear repression of the transcription activation by p73 by this Ad early protein was observed. In addition, we found that stable expression of the Ad5 E1A and that of the E1B protein both caused increased p73 protein expression. The large E1B and the E4orf6 proteins together do not target the p73 protein for rapid degradation after adenoviral infection, as has previously been found for the p53 protein, probably because the large E1B protein does not interact with p73. Our results suggest that the p53 and p73 proteins are both inactivated after Ad infection and transformation but via distinct mechanisms.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/genética , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Regulação da Expressão Gênica , Genes Reporter/genética , Genes Supressores de Tumor , Humanos , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional/genética , Transformação Genética , Células Tumorais Cultivadas , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor
10.
Mol Cell Biol ; 21(23): 8225-35, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689711

RESUMO

AFX is a Forkhead transcription factor that induces a G(1) cell cycle arrest via upregulation of the cell cycle inhibitor p27(Kip1). Previously we have shown that protein kinase B (PKB) phosphorylates AFX causing inhibition of AFX by nuclear exclusion. In addition, Ras, through the activation of the RalGEF-Ral pathway, induces phosphorylation of AFX. Here we show that the Ras-Ral pathway provokes phosphorylation of threonines 447 and 451 in the C terminus of AFX. A mutant protein in which both threonines are substituted for alanines (T447A/T451A) still responds to PKB-regulated nuclear-cytoplasmic shuttling, but transcriptional activity and consequent G(1) cell cycle arrest are greatly impaired. Furthermore, inhibition of the Ral signaling pathway abolishes both AFX-mediated transcription and regulation of p27(Kip1), while activation of Ral augments AFX activity. From these results we conclude that Ral-mediated phosphorylation of threonines 447 and 451 is required for proper activity of AFX-WT. Interestingly, the T447A/T451A mutation did not affect the induction of transcription and G(1) cell cycle arrest by the PKB-insensitive AFX-A3 mutant, suggesting that Ral-mediated phosphorylation plays a role in the regulation of AFX by PKB.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Serina-Treonina Quinases , Fatores de Transcrição/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Células 3T3 , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Ativação Enzimática/fisiologia , Fatores de Transcrição Forkhead , Genes Reporter , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Treonina/metabolismo , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Proteínas ras/metabolismo
11.
Mol Cell Biol ; 20(22): 8480-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11046144

RESUMO

The transcription factor c-Jun is critically involved in the regulation of proliferation and differentiation as well as cellular transformation induced by oncogenic Ras. The signal transduction pathways that couple Ras activation to c-Jun phosphorylation are still partially elusive. Here we show that an activated version of the Ras effector Rlf, a guanine nucleotide exchange factor (GEF) of the small GTPase Ral, can induce the phosphorylation of serines 63 and 73 of c-Jun. In addition, we show that growth factor-induced, Ras-mediated phosphorylation of c-Jun is abolished by inhibitory mutants of the RalGEF-Ral pathway. These results suggest that the RalGEF-Ral pathway plays a major role in Ras-dependent c-Jun phosphorylation. Ral-dependent regulation of c-Jun phosphorylation includes JNK, a still elusive JNKK, and possibly Src.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Proteínas ras/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina , Humanos , Insulina/metabolismo , Insulina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase 4 , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Serina , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
12.
Mol Cell Biol ; 9(10): 4312-22, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2555688

RESUMO

Expression of a mutant H-ras gene confers a transformed phenotype to rat-1 fibroblasts which is basically independent of exogenous growth factors (GFs). Rat-1 cells induced to express high levels of the normal H-ras gene were also found to display a transformed phenotype. In contrast to cells expressing mutant H-ras, these cells were dependent on GFs. We used this difference in GF dependence to analyze a possible involvement of exogenous GFs in H-ras function. Compared with untransformed rat-1 cells, cells overexpressing normal H-ras displayed an elevated response toward insulinlike growth factor 1 (IGF-1), insulin, and bombesin and an increased sensitivity toward phosphatidic acids. It was found that 8-bromo-cyclic AMP inhibited the responses to all GFs in rat-1 cells but had no effect on mutant-H-ras-transformed cells. In cells overexpressing normal H-ras, 8-bromo-cyclic AMP inhibited the responses to all GFs except those to insulin and IGF-1. This implies that overexpression of normal H-ras in the presence of insulin/IGF-1 is functionally similar to the expression of mutant H-ras, since mutant H-ras can circumvent this block by itself. These and other results strongly suggest a functional linkage between insulin/IGF-1 and normal p21 H-ras.


Assuntos
Substâncias de Crescimento/fisiologia , Proteína Oncogênica p21(ras)/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica/metabolismo , AMP Cíclico/fisiologia , Replicação do DNA/efeitos dos fármacos , Expressão Gênica , Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Fosforilação , Receptor de Insulina/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Somatomedina , Transdução de Sinais/fisiologia
13.
Mol Cell Biol ; 12(8): 3425-30, 1992 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1321335

RESUMO

The p21ras GTPase-activating protein (GAP) is thought to function as both a negative regulator and a downstream target of p21ras. Here, we have investigated the role of GAP by using a transient expression assay with a fos luciferase reporter plasmid. We used GAP deletion mutants that lack the domain involved in interaction with p21ras and encode essentially only the SH2-SH3 domains. When these GAP deletion mutants were expressed, we observed a marked induction of fos promoter activity similar to induction by activated p21ras. Expression of a full-length GAP construct had no effect on the activity of the fos promoter. Activation of the fos promoter by these GAP SH2-SH3 regions was inhibited by cotransfection of a dominant inhibitory mutant of p21ras, Ras(Asn-17). Thus, the induction of gene expression by GAP SH2-SH3 domains is dependent on p21ras activity. Moreover, induction of fos promoter activity by GAP SH2-SH3 domains is increased severalfold after cotransfection of an activated mutant of p21ras, Ras(Leu-61), or insulin stimulation of A14 cells, both leading to an increase in the levels of GTP-bound p21ras. The combined effect of Ras(Leu-61) and the GAP deletion mutants was not inhibited by Ras(Asn-17), indicating that GAP SH2-SH3 domains do not function to activate endogenous p21ras but cooperate with another signal coming from active p21ras. These data suggest that GAP SH2-SH3 domains serve to induce gene expression by p21ras but that additional signals coming from p21ras are required for them to function.


Assuntos
Regulação da Expressão Gênica , Genes ras , Proteínas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Células 3T3 , Animais , Vírus do Sarcoma Aviário/genética , Células CHO , Deleção Cromossômica , Células Clonais , Cricetinae , Proteínas Ativadoras de GTPase , Insulina/farmacologia , Cinética , Luciferases/genética , Luciferases/metabolismo , Camundongos , Plasmídeos , Regiões Promotoras Genéticas , Proteínas/genética , Transfecção , Proteínas Ativadoras de ras GTPase
14.
Mol Cell Biol ; 18(4): 1802-11, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528752

RESUMO

Regulation of phosphoinositide 3-kinase (PI 3-kinase) can occur by binding of the regulatory p85 subunit to tyrosine-phosphorylated proteins and by binding of the p110 catalytic subunit to activated Ras. However, the way in which these regulatory mechanisms act to regulate PI 3-kinase in vivo is unclear. Here we show that several growth factors (basic fibroblast growth factor [bFGF], platelet-derived growth factor [PDGF], and epidermal growth factor [EGF; to activate an EGF receptor-Ret chimeric receptor]) all activate PI 3-kinase in vivo in the neuroectoderm-derived cell line SKF5. However, these growth factors differ in their ability to activate PI 3-kinase-dependent signaling. PDGF and EGF(Ret) treatment induced PI 3-kinase-dependent lamellipodium formation and protein kinase B (PKB) activation. In contrast, bFGF did not induce lamellipodium formation but activated PKB, albeit to a small extent. PDGF and EGF(Ret) stimulation resulted in binding of p85 to tyrosine-phosphorylated proteins and strong Ras activation. bFGF, however, induced only strong activation of Ras. In addition, while RasAsn17 abolished bFGF activation of PKB, PDGF- and EGF(Ret)-induced PKB activation was only partially inhibited and lamellipodium formation was unaffected. Interestingly, in contrast to activation of only endogenous Ras (bFGF), ectopic expression of activated Ras did result in lamellipodium formation. From this we conclude that, in vivo, p85 and Ras synergize to activate PI 3-kinase and that strong activation of only endogenous Ras exerts a small effect on PI 3-kinase activity, sufficient for PKB activation but not lamellipodium formation. This differential sensitivity to PI 3-kinase activation could be explained by our finding that PKB activation and lamellipodium formation are independent PI 3-kinase-induced events.


Assuntos
Movimento Celular , Proteínas de Drosophila , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas ras/metabolismo , Androstadienos/farmacologia , Cromonas/farmacologia , Citoplasma/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Tirosina/metabolismo , Wortmanina
15.
Mol Cell Biol ; 14(3): 1575-81, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8114695

RESUMO

Shc proteins are phosphorylated on tyrosine residues and associate with growth factor receptor-bound protein 2 (Grb2) upon treatment of cells with epidermal growth factor (EGF) or insulin. We have studied the role of Shc in insulin- and EGF-induced activation of p21ras in NIH 3T3 cells overexpressing human insulin receptors (A14 cells). A14 cells are equally responsive to insulin and EGF with respect to activation of p21ras. Analysis of Shc immunoprecipitates revealed that (i) both insulin and EGF treatment resulted in Shc tyrosine phosphorylation and (ii) Shc antibodies coimmunoprecipitated both Grb2 and mSOS after insulin and EGF treatment. The induction of tyrosine phosphorylation of Shc and the presence of Grb2 and mSOS in Shc immunoprecipitates followed similar time courses, with somewhat higher levels after EGF treatment. In mSOS immunoprecipitates, Shc could be detected as well. Furthermore, Shc immune complexes contained guanine nucleotide exchange activity toward p21ras in vitro. From these results, we conclude that after insulin and EGF treatment, Shc associates with both Grb2 and mSOS and therefore may mediate, at least in part, insulin- and EGF-induced activation of p21ras. In addition, we investigated whether the Grb2-mSOS complex associates with the insulin receptor or with insulin receptor substrate 1 (IRS1). Although we observed association of Grb2 with IRS1, we did not detect complex formation between mSOS and IRS1 in experiments in which the association of mSOS with Shc was readily detectable. Furthermore, whereas EGF treatment resulted in the association of mSOS with the EGF receptor, insulin treatment did not result in the association of mSOS with the insulin receptor. These results indicate that the association of Grb2-nSOS with Shc may be an important event in insulin-induced, mSOS-mediated activation of p21ras.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Receptores ErbB/fisiologia , Proteínas/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Linhagem Celular , Fator de Crescimento Epidérmico/farmacologia , Proteína Adaptadora GRB2 , Nucleotídeos de Guanina/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteínas Son Of Sevenless , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Proteínas Virais/metabolismo
16.
Mol Cell Biol ; 13(12): 7248-56, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8246947

RESUMO

Expression of p21rasAsn-17, a dominant negative mutant of p21ras that blocks p21ras activation by growth factors, inhibits activation of extracellular signal-regulated kinase 2 (ERK2) by insulin and platelet-derived growth factor in rat-1 cells [A. M. M. de Vries-Smits, B. M. T. Burgering, S. J. Leevers, C. J. Marshall, and J. L. Bos, Nature (London) 357:602-604, 1992]. Here we report that expression of p21rasAsn-17 does not abolish epidermal growth factor (EGF)-induced phosphorylation of ERK2 in fibroblasts. Since EGF activates p21ras in these cells, this indicates that EGF induces a p21ras-independent pathway for the phosphorylation of ERK2 as well. We investigated whether activation of protein kinase C (PKC) or increase in intracellular calcium could be involved in p21ras-independent signaling. In rat-1 cells, inhibition of either PKC, by prolonged 12-O-tetradecanoylphorbol-13-acetate (TPA) pretreatment, or calcium influx, by ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) pretreatment, did not abolish EGF-induced ERK2 phosphorylation. However, a combined inhibition of both p21ras and calcium influx, but not PKC, resulted in a complete inhibition of EGF-induced ERK2 phosphorylation. In contrast, in Swiss 3T3 cells, inhibition of both p21ras activation and TPA-sensitive PKC, but not calcium influx, inhibited EGF-induced ERK2 phosphorylation. These results demonstrate that in fibroblasts, EGF induces alternative pathways of ERK2 phosphorylation in a cell-type-specific manner.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Ácido Egtázico/farmacologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
17.
Mol Cell Biol ; 13(1): 155-62, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8417322

RESUMO

A number of growth factors, including insulin and epidermal growth factor (EGF), induce accumulation of the GTP-bound form of p21ras. This accumulation could be caused either by an increase in guanine nucleotide exchange on p21ras or by a decrease in the GTPase activity of p21ras. To investigate whether insulin and EGF affect nucleotide exchange on p21ras, we measured binding of [alpha-32P]GTP to p21ras in cells permeabilized with streptolysin O. For this purpose, we used a cell line which expressed elevated levels of p21 H-ras and which was highly responsive to insulin and EGF. Stimulation with insulin or EGF resulted in an increase in the rate of nucleotide binding to p21ras. To determine whether this increased binding rate is due to the activation of a guanine nucleotide exchange factor, we made use of the inhibitory properties of a dominant negative mutant of p21ras, p21ras (Asn-17). Activation of p21ras by insulin and EGF in intact cells was abolished in cells infected with a recombinant vaccinia virus expressing p21ras (Asn-17). In addition, the enhanced nucleotide binding to p21ras in response to insulin and EGF in permeabilized cells was blocked upon expression of p21ras (Asn-17). From these data, we conclude that the activation of a guanine nucleotide exchange factor is involved in insulin- and EGF-induced activation of p21ras.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Nucleotídeos de Guanina/metabolismo , Insulina/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Células Cultivadas , Técnicas In Vitro , Proteínas Proto-Oncogênicas p21(ras)/química , Ratos , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
18.
Mol Cell Biol ; 20(3): 779-85, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10629034

RESUMO

Rap1, a small GTPase of the Ras family, is ubiquitously expressed and particularly abundant in platelets. Previously we have shown that Rap1 is rapidly activated after stimulation of human platelets with alpha-thrombin. For this activation, a phospholipase C-mediated increase in intracellular calcium is necessary and sufficient. Here we show that thrombin induces a second phase of Rap1 activation, which is mediated by protein kinase C (PKC). Indeed, the PKC activator phorbol 12-myristate 13-acetate induced Rap1 activation, whereas the PKC-inhibitor bisindolylmaleimide inhibited the second, but not the first, phase of Rap1 activation. Activation of the integrin alpha(IIb)beta(3), a downstream target of PKC, with monoclonal antibody LIBS-6 also induced Rap1 activation. However, studies with alpha(IIb)beta(3)-deficient platelets from patients with Glanzmann's thrombasthenia type 1 show that alpha(IIb)beta(3) is not essential for Rap1 activation. Interestingly, induction of platelet aggregation by thrombin resulted in the inhibition of Rap1 activation. This downregulation correlated with the translocation of Rap1 to the Triton X-100-insoluble, cytoskeletal fraction. We conclude that in platelets, alpha-thrombin induces Rap1 activation first by a calcium-mediated pathway independently of PKC and then by a second activation phase mediated by PKC and, in part, integrin alpha(IIb)beta(3). Inactivation of Rap1 is mediated by an aggregation-dependent process that correlates with the translocation of Rap1 to the cytoskeletal fraction.


Assuntos
Plaquetas/metabolismo , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Trombastenia/sangue , Proteínas rap1 de Ligação ao GTP/sangue , Anticorpos Monoclonais/farmacologia , Plaquetas/efeitos dos fármacos , Cálcio/sangue , Citoesqueleto/metabolismo , Humanos , Técnicas In Vitro , Indóis/farmacologia , Maleimidas/farmacologia , Fator de Ativação de Plaquetas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/sangue , Valores de Referência , Acetato de Tetradecanoilforbol/farmacologia , Trombina/farmacologia , Fosfolipases Tipo C/sangue , Proteínas rap1 de Ligação ao GTP/biossíntese
19.
Mol Cell Biol ; 18(5): 2486-91, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9566869

RESUMO

Ral is a ubiquitously expressed Ras-like small GTPase which is abundantly present in human platelets. The biological function of Ral and the signaling pathway in which Ral is involved are largely unknown. Here we describe a novel method to measure Ral activation utilizing the Ral binding domain of the putative Ral effector RLIP76 as an activation-specific probe. With this assay we investigated the signaling pathway that leads to Ral activation in human platelets. We found that Ral is rapidly activated after stimulation with various platelet agonists, including alpha-thrombin. In contrast, the platelet antagonist prostaglandin I2 inhibited alpha-thrombin-induced Ral activation. Activation of Ral by alpha-thrombin could be inhibited by depletion of intracellular Ca2+, whereas the induction of intracellular Ca2+ resulted in the activation of Ral. Our results show that Ral can be activated by extracellular stimuli. Furthermore, we show that increased levels of intracellular Ca2+ are sufficient for Ral activation in platelets. This activation mechanism correlates with the activation mechanism of the small GTPase Rap1, a putative upstream regulator of Ral guanine nucleotide exchange factors.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Plaquetas/enzimologia , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase , Ativação Plaquetária/fisiologia , Cálcio/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ativação Enzimática , Epoprostenol/farmacologia , Guanosina Trifosfato/metabolismo , Humanos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Ligação Proteica , Transdução de Sinais , Trombina/farmacologia , Proteínas ral de Ligação ao GTP , Proteínas rap de Ligação ao GTP
20.
Mol Cell Biol ; 10(11): 5857-64, 1990 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2172787

RESUMO

The adenovirus early region 1A (E1A) oncogene interferes with the expression level and activity of the AP-1 transcription factor family. E1A abolished the transactivating function of AP-1 (Jun/Fos), which binds to the 12-O-tetradecanoylphorbol-13-acetate-responsive element of the collagenase gene (collTRE). In contrast, the activity of another member of the AP-1 family that binds to the c-junTRE was not repressed. The mRNA expression of the c-jun gene was, in fact, strongly elevated in various cell types expressing the E1A gene of either adenovirus type 5 (Ad5) or Ad12. The regulation of the junB gene by adenovirus E1A, on the other hand, depended both on the cell type and on the transforming adenovirus serotype. The fact that E1A-induced alterations in the repertoire of AP-1 transcription factors depend on its transforming domain in conserved region 1 suggests that the effects are relevant for the transformation process.


Assuntos
Adenovírus Humanos/genética , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/genética , Proteínas Oncogênicas Virais/genética , Oncogenes , Fatores de Transcrição/genética , Proteínas Precoces de Adenovirus , Animais , Linhagem Celular , Humanos , Mutação , Polyomavirus/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun , Proto-Oncogenes , RNA Mensageiro/genética , Vírus 40 dos Símios/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa