Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Curr Opin Cell Biol ; 6(2): 204-11, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8024811

RESUMO

The past year has seen remarkable advances in our understanding of how members of the Ras superfamily of GTPases are activated. Not only have new guanine-nucleotide exchange factors (GEFs) that activate these GTPases been identified, but in some cases, the mechanisms by which GEFs are connected to events at the cell surface have also been described. We have also come to appreciate the possibilities that GEFs may be multifunctional and that they may contribute to tissue-specific regulation of their cognate GTPase.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinais , Animais , Membrana Celular/metabolismo , GTP Fosfo-Hidrolases/química , Proteínas de Ligação ao GTP/química , Humanos , Modelos Biológicos , Modelos Estruturais
2.
Nat Cell Biol ; 1(2): E25-7, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10559887

RESUMO

One of the most powerful ways of studying the function of a protein is to specifically block its activity within cells. Over the past decade, dominant-inhibitory proteins have emerged as popular tools with which to accomplish this task; these mutated proteins interfere with the function of their normal cellular counterparts or with proteins that interact with them. This approach has been used extensively in the elucidation of signal-transduction cascades, such as those involving Ras-family proteins. Here I discuss the power and potential pitfalls of using dominant-inhibitory Ras proteins.


Assuntos
Fenômenos Fisiológicos Celulares , Proteínas de Ligação ao GTP/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Mutagênese , Animais , Sítios de Ligação , Proteínas de Ligação ao GTP/antagonistas & inibidores , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais
3.
J Cell Biol ; 97(5 Pt 1): 1435-43, 1983 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-6630290

RESUMO

A mitogenic polypeptide, previously identified in Sertoli cells of the prepuberal mouse (Feig, L. A., A. R. Bellvé, N. Horbach-Erickson, and M. Klagsbrun, 1980, Proc. Natl. Acad. Sci. USA., 77:4774-4778), now has been shown to exist in Sertoli cells of the adult mouse and in the seminiferous epithelium of several other mammalian species, including the rat, guinea pig, and calf. The levels of this seminiferous growth factor (SGF) are not appreciably reduced in adult mouse testes following hypophysectomy. SGF purified from either the adult mouse or newborn calf seminiferous epithelium has a molecular weight (Mr) of 15,700 and a pl between 4.8 and 5.8, when exposed to denaturing conditions. Furthermore, SGF from these two mammalian species probably has few exposed hydrophobic domains and has a strong propensity to aggregate into multiple, high Mr species. A purification sequence based on these biochemical properties has enabled a greater than 350-fold enrichment of SGF activity from the calf seminiferous epithelium. The protocol involves a sequence of: (a) ammonium sulfate precipitation, (b) DEAE-cellulose ion exchange chromatography, (c) gel filtration chromatography on Bio-Gel P150 in 1.0 M ammonium acetate, (d) hydrophobic chromatography on dodecyl agarose, and (e) gel filtration chromatography in 6.0 M guanidine hydrochloride. Subsequent analysis of this purified preparation by SDS PAGE, followed by silver staining, reveals approximately 7 polypeptides with Mr between 14,000 and 20,000.


Assuntos
Mitógenos/isolamento & purificação , Túbulos Seminíferos/análise , Testículo/análise , Animais , Bovinos , DNA/metabolismo , Epitélio/análise , Fibroblastos/efeitos dos fármacos , Cobaias , Hipofisectomia , Ponto Isoelétrico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Timidina/metabolismo
4.
J Cell Biol ; 130(3): 725-31, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7622571

RESUMO

The PDGF receptor-beta mediates both mitogenic and chemotactic responses to PDGF-BB. Although the role of Ras in tyrosine kinase-mediated mitogenesis has been characterized extensively, its role in PDGF-stimulated chemotaxis has not been defined. Using cells expressing a dominant-negative ras, we find that Ras inhibition suppresses migration toward PDGF-BB. Overexpression of either Ras-GTPase activating protein (Ras-GAP) or a Ras guanine releasing factor (GRF) also inhibited PDGF-stimulated chemotaxis. In addition, cells producing excess constitutively active Ras failed to migrate toward PDGF-BB, consistent with the observation that either excess ligand or excess signaling intermediate can suppress the chemotactic response. These results suggest that Ras can function in normal cells to support chemotaxis toward PDGF-BB and that either too little or too much Ras activity can abrogate the chemotactic response. In contrast to Ras overexpression, cells producing excess constitutively active Raf, a downstream effector of Ras, did migrate toward PDGF-BB. Cells expressing dominant-negative Ras were able to migrate toward soluble fibronectin demonstrating that these cells retained the ability to migrate. These results suggest that Ras is an intermediate in PDGF-stimulated chemotaxis but may not be required for fibronectin-stimulated cell motility.


Assuntos
Quimiotaxia/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Células 3T3 , Animais , Becaplermina , Western Blotting , Fibronectinas/farmacologia , Proteínas Ativadoras de GTPase , Fatores de Troca do Nucleotídeo Guanina , Lisofosfolipídeos/farmacologia , Camundongos , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-raf , Proteínas Proto-Oncogênicas c-sis , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Ativadoras de ras GTPase , Fatores ras de Troca de Nucleotídeo Guanina , ras-GRF1
5.
Science ; 223(4637): 698-701, 1984 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-6695178

RESUMO

A tumor isolate from a patient with serous cystadenocarcinoma of the ovary contained an activated rasK gene detected hy transfection of NIH/3T3 cells. In contrast, DNA from normal cells of the same patient lacked transforming activity, indicating that activation of this transforming gene was the consequence of somatic mutation in the neoplastic cells. The transforming gene product displayed an electrophoretic mobility in sodium dodecyl sulfate-polyacrylamide gels that differed from the mobilities of rasK transforming proteins in other tumors, indicating that a previously undescribed mutation was responsible for activation of rasK in this ovarian carcinoma.


Assuntos
Cistadenocarcinoma/genética , Oncogenes , Neoplasias Ovarianas/genética , Animais , Sequência de Bases , Linhagem Celular , Transformação Celular Neoplásica , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Feminino , Humanos , Neoplasias Pulmonares/genética , Camundongos , Transfecção
6.
Trends Biochem Sci ; 21(11): 438-41, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8987400

RESUMO

It is becoming clear that Ras proteins mediate their diverse biological functions by binding to, and participating in, the activation of multiple downstream targets. Recent work has identified nucleotide-exchange factors for Ral-GTPases as the newest members of the set of putative Ras 'effector molecules'. This new work has also detected two potential downstream targets of Ral proteins, a novel CDC42/Rac GTPase-activating protein and a phospholipase D.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Fosfolipase D/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo , Animais , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP , Proteínas rac de Ligação ao GTP , Proteínas rap de Ligação ao GTP
7.
Mol Cell Biol ; 11(10): 4822-9, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1922022

RESUMO

We have previously demonstrated that substitution of Asn for Ser at position 17 of RasH yields a dominant inhibitory protein whose expression in cells interferes with endogenous Ras function (L. A. Feig, and G. M. Cooper, Mol. Cell. Biol. 8:3235-3243, 1988). Subsequent structural studies have shown that the hydroxyl group of Ser-17 contributes to the binding of Mg2+ associated with bound nucleotide. In this report, we show that more subtle amino acid substitutions at this site that would be expected to interfere with complexing Mg2+, such as Cys or Ala, also generated dominant inhibitory mutants. In contrast, a Thr substitution that conserves a reactive hydroxyl group maintained normal Ras function. These results argue that the defect responsible for the inhibitory activity is improper coordination of Mg2+. Preferential affinity for GDP, observed in the original Asn-17 mutant, was found exclusively in inhibitory mutants. However, this binding specificity did not completely block the mutant proteins from binding GTP in vivo since introduction of the autophosphorylation site, Thr-59, in 17N Ras resulted in the phosphorylation of the double mutant in cells. Furthermore, inhibitory mutants failed to activate a model downstream target, yeast adenylate cyclase, even when bound to GTP. Thus, the consequence of improper complexing of Mg2+ was to lock the protein in a constitutively inactive state. A model is presented to explain how these properties could cause the mutant protein to inhibit the activation of endogenous Ras by competing for a guanine nucleotide-releasing factor.


Assuntos
Guanosina Trifosfato/metabolismo , Magnésio/metabolismo , Mutação , Proteína Oncogênica p21(ras)/metabolismo , Adenilil Ciclases/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Modelos Biológicos , Proteína Oncogênica p21(ras)/genética , Fosforilação , Testes de Precipitina , Ratos
8.
Mol Cell Biol ; 8(6): 2472-8, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3043178

RESUMO

The effect of a series of mutations on the transforming potential of normal human rasH has been compared with their effects on GTPase and guanine nucleotide exchange rates of p21. The mutation Val-146 resulted in partial activation of transforming potential which could be attributed to a greater than 1,000-fold-increased rate of nucleotide exchange in the absence of an effect on GTPase. In contrast, the more modest enhancement of exchange rate (approximately 100-fold) which resulted from the mutation Met-14 did not affect biological activity. The partially activating mutation Thr-59 was found to result in both a 5-fold reduction in GTPase and a 10-fold increase in nucleotide exchange. However, the nontransforming mutant Ile-59 displayed a comparable decrease in GTPase without an effect on nucleotide exchange. The activating effect of the Thr-59 mutation may thus represent a combined effect of reduced GTPase and increased exchange. Similarly, the strongly activating mutation Leu-61 resulted in a fivefold increase in nucleotide exchange in addition to decreased GTPase, whereas weakly activating mutations at position 61 (Trp and Pro) resulted only in decreased GTPase without affecting nucleotide exchange rates. Finally, combining the two mutations Met-14 and Ile-59, which alone had no effect on biological activity, yielded a double mutant with a 20-fold increased transforming potential, demonstrating a synergistic effect of these two mutations. Overall, these results indicate that large increases in nucleotide exchange can activate ras transforming potential in the absence of decreased GTPase and that relatively modest increases in nucleotide exchange can act synergistically with decreased GTPase to contribute to ras activation.


Assuntos
Proteínas de Membrana/metabolismo , Mutação , Proteínas Proto-Oncogênicas/metabolismo , Fibroblastos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Transfecção
9.
Mol Cell Biol ; 8(8): 3235-43, 1988 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3145408

RESUMO

Substitution of asparagine for serine at position 17 decreased the affinity of rasH p21 for GTP 20- to 40-fold without significantly affecting its affinity for GDP. Transfection of NIH 3T3 cells with a mammalian expression vector containing the Asn-17 rasH gene and a Neor gene under the control of the same promoter yielded only a small fraction of the expected number of G418-resistant colonies, indicating that expression of Asn-17 p21 inhibited cell proliferation. The inhibitory effect of Asn-17 p21 required its localization to the plasma membrane and was reversed by coexpression of an activated ras gene, indicating that the mutant p21 blocked the endogenous ras function required for NIH 3T3 cell proliferation. NIH 3T3 cells transformed by v-mos and v-raf, but not v-src, were resistant to inhibition by Asn-17 p21, indicating that the requirement for normal ras function can be bypassed by these cytoplasmic oncogenes. The Asn-17 mutant represents a novel reagent for the study of ras function by virtue of its ability to inhibit cellular ras activity in vivo. Since this phenotype is likely associated with the preferential affinity of the mutant protein for GDP, analogous mutations might also yield inhibitors of other proteins whose activities are regulated by guanine nucleotide binding.


Assuntos
Divisão Celular , Nucleotídeos de Guanina/metabolismo , Guanosina Difosfato/metabolismo , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Animais , Asparagina , Células Cultivadas , Clonagem Molecular , Guanosina Trifosfato/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos , Mutação , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas p21(ras) , Serina , Transfecção
10.
Mol Cell Biol ; 15(8): 4578-84, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7623849

RESUMO

Ral proteins constitute a distinct family of Ras-related GTPases. Although similar to Ras in amino acid sequence, Ral proteins are activated by a unique nucleotide exchange factor and inactivated by a distinct GTPase-activating protein. Unlike Ras, they fail to promote transformed foci when activated versions are expressed in cells. To identify downstream targets that might mediate a Ral-specific function, we used a Saccharomyces cerevisiae-based interaction assay to clone a novel cDNA that encodes a Ral-binding protein (RalBP1). RalBP1 binds specifically to the active GTP-bound form of RalA and not to a mutant Ral with a point mutation in its putative effector domain. In addition to a Ral-binding domain, RalBP1 also contains a Rho-GTPase-activating protein domain that interacts preferentially with Rho family member CDC42. Since CDC42 has been implicated in bud site selection in S. cerevisiae and filopodium formation in mammalian cells, Ral may function to modulate the actin cytoskeleton through its interactions with RalBP1.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Fúngicas/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase , Transdução de Sinais , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , DNA Complementar/genética , Proteínas Fúngicas/genética , Biblioteca Gênica , Immunoblotting , Dados de Sequência Molecular , Ligação Proteica , Ratos , Proteínas Recombinantes/biossíntese , Saccharomyces cerevisiae/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP , Proteínas ral de Ligação ao GTP
11.
Mol Cell Biol ; 19(3): 1731-41, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10022860

RESUMO

Ras proteins can activate at least three classes of downstream target proteins: Raf kinases, phosphatidylinositol-3 phosphate (PI3) kinase, and Ral-specific guanine nucleotide exchange factors (Ral-GEFs). In NIH 3T3 cells, activated Ral-GEFs contribute to Ras-induced cell proliferation and oncogenic transformation by complementing the activities of Raf and PI3 kinases. In PC12 cells, activated Raf and PI3 kinases mediate Ras-induced cell cycle arrest and differentiation into a neuronal phenotype. Here, we show that in PC12 cells, Ral-GEF activity acts opposite to other Ras effectors. Elevation of Ral-GEF activity induced by transfection of a mutant Ras protein that preferentially activates Ral-GEFs, or by transfection of the catalytic domain of the Ral-GEF Rgr, suppressed cell cycle arrest and neurite outgrowth induced by nerve growth factor (NGF) treatment. In addition, Rgr reduced neurite outgrowth induced by a mutant Ras protein that preferentially activates Raf kinases. Furthermore, inhibition of Ral-GEF activity by expression of a dominant negative Ral mutant accelerated cell cycle arrest and enhanced neurite outgrowth in response to NGF treatment. Ral-GEF activity may function, at least in part, through inhibition of the Rho family GTPases, CDC42 and Rac. In contrast to Ras, which was activated for hours by NGF treatment, Ral was activated for only approximately 20 min. These findings suggest that one function of Ral-GEF signaling induced by NGF is to delay the onset of cell cycle arrest and neurite outgrowth induced by other Ras effectors. They also demonstrate that Ras has the potential to promote both antidifferentiation and prodifferentiation signaling pathways through activation of distinct effector proteins. Thus, in some cell types the ratio of activities among Ras effectors and their temporal regulation may be important determinants for cell fate decisions between proliferation and differentiation.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/metabolismo , Neuritos/fisiologia , Proteínas/metabolismo , Animais , Ciclo Celular , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Genes fos , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Fatores de Crescimento Neural/farmacologia , Células PC12 , Regiões Promotoras Genéticas , Coelhos , Ratos , Proteínas ral de Ligação ao GTP , Fatores ras de Troca de Nucleotídeo Guanina , Proteínas ras/genética , Proteínas ras/metabolismo , Proteínas rho de Ligação ao GTP
12.
Mol Cell Biol ; 11(8): 4053-64, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2072908

RESUMO

Two dominant inhibitory Ras mutant proteins were analyzed by microinjection. One, [Asn-17]Ras, had a substitution in the putative Mg(2+)-binding site of Ha-Ras. The other, RAST, had a mutation in a yeast RAS protein that impaired its GTPase activity and increased its affinity for GAP. RAST also had a mutation that blocked its localization to the plasma membrane. In NIH 3T3 cells [Asn-17]Ras inhibited the function of normal Ras much more efficiently than that of oncogenic Ras. In contrast, RAST interfered with the transforming activity of oncogenic Ras more efficiently than that of normal Ras. These conclusions were based on two separate types of analysis. The inhibitory Ras mutant proteins were first microinjected into cells stably transformed either by oncogenic Ras or by high levels of expression of cellular Ras. Results obtained in stably transformed cells were then verified by coinjection of the inhibitory Ras mutant proteins together with transforming concentrations of either oncogenic or normal Ras protein. Whereas RAST was active in soluble form. [Asn-17]Ras required membrane localization for activity. Furthermore, mutations in the GAP/effector-binding domain reduced or eliminated the inhibitory activity of RAST but had no detectable effect on [Asn-17]Ras. These results are consistent with the possibility that [Asn-17]Ras functions by blocking the activation of endogenous Ras proteins, while RAST functions by blocking the ability of activated Ras to stimulate a downstream target within the cells. The properties of RAST suggest that interference with the GAP/effector-binding function of RAS represents a strategy for the preferential inactivation of oncogenic Ras in cells.


Assuntos
Transformação Celular Neoplásica , Genes Dominantes , Genes ras , Proteína Oncogênica p21(ras)/genética , Animais , Divisão Celular , Linhagem Celular , Replicação do DNA , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Camundongos , Microinjeções , Proteína Oncogênica p21(ras)/metabolismo , Proteínas Recombinantes/metabolismo , Timidina/metabolismo
13.
Mol Cell Biol ; 21(8): 2650-8, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11283245

RESUMO

Ras proteins have the capacity to bind to and activate at least three families of downstream target proteins: Raf kinases, phosphatidylinositol 3 (PI 3)-kinase, and Ral-specific guanine nucleotide exchange factors (Ral-GEFs). We have previously shown that the Ras/Ral-GEF and Ras/Raf pathways oppose each other upon nerve growth factor stimulation, with the former promoting proliferation and the latter promoting cell cycle arrest. Moreover, the pathways are not activated equally. While the Ras/Raf/Erk signaling pathway is induced for hours, the Ras/Ral-GEF/Ral signaling pathway is induced for only minutes. Here we show that this preferential down-regulation of Ral signaling is mediated, at least in part, by protein kinase C (PKC). In particular, we show that PKC activation by phorbol ester treatment of cells blocks growth factor-induced Ral activation while it enhances Erk activation. Moreover, suppression of growth factor-induced PKC activation enhances and prolongs Ral activation. PKC does not influence the basal activity of the Ral-GEF designated Ral-GDS but suppresses its activation by Ras. Interestingly, Ras binding to the C-terminal Ras binding domain of Ral-GDS is not affected by PKC activity. Instead, suppression of Ral-GDS activation occurs through the region N terminal to the catalytic domain, which becomes phosphorylated in response to phorbol ester treatment of cells. These findings identify a role for PKC in determining the specificity of Ras signaling by its ability to differentially modulate Ras effector protein activation.


Assuntos
Proteína Quinase C/metabolismo , Proteínas ras/metabolismo , Animais , Células COS , Ativação Enzimática/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Neural/farmacologia , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Deleção de Sequência , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia , Fator ral de Troca do Nucleotídeo Guanina/química , Fator ral de Troca do Nucleotídeo Guanina/genética , Fator ral de Troca do Nucleotídeo Guanina/metabolismo
14.
Mol Cell Biol ; 11(10): 4830-8, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1656212

RESUMO

The properties of a Gs alpha mutant with an Asn substituted for Ser at position 54, designated mutant 54Asn alpha s, were studied after expression in S49 alpha s-deficient (cyc-) cells. Ser-54 in alpha s is comparable to Ser-17 in Ras, which is involved in binding Mg2+ associated with bound nucleotide. 54Asn alpha s did not restore either hormone-induced cyclic AMP production in intact cyc- cells or hormone-induced adenylyl cyclase activation in membranes isolated from these cells. The defect was a failure of ligand-bound receptor to activate 54Asn alpha s, since the mutant protein retained the ability to activate adenylyl cyclase in isolated membranes in the presence of GTP or GTP gamma S. Guanine nucleotide regulation of mutant alpha s suggested that it has increased guanine nucleotide exchange rates and an increased preference for diphosphates over triphosphates. Hormone stimulation magnified the preference of 54Asn alpha s for diphosphates, which could account for its inability to be activated by receptor. The properties of this mutant are discussed in terms of similarities to and differences with the analogous RasH mutant, which has been shown to interfere with endogenous Ras function in cells.


Assuntos
Proteínas de Ligação ao GTP/genética , Magnésio/metabolismo , Mutação/genética , Adenilil Ciclases/metabolismo , Animais , Asparagina/genética , Sítios de Ligação/genética , Northern Blotting , AMP Cíclico/metabolismo , Ativação Enzimática , Proteínas de Ligação ao GTP/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Trifosfato/metabolismo , Isoproterenol/farmacologia , Cinética , Camundongos , Proteína Oncogênica p21(ras)/genética , Serina/genética , Células Tumorais Cultivadas
15.
Mol Cell Biol ; 16(9): 4888-96, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8756648

RESUMO

We have recently shown that the neuronal exchange factor p140 Ras-GRF becomes activated in vivo in response to elevated calcium levels [C. L. Farnsworth, N. W. Freshney, L. B. Rosen, A. Ghosh, M. E. Greenberg, and L. A. Feig, Nature (London) 376:524-527, 1995]. Activation is mediated by calcium-induced calmodulin binding to an IQ domain near the N terminus of Ras-GRF. Here we show that the adjacent N-terminal pleckstrin homology (PH), coiled-coil, and IQ domains function cooperatively to allow Ras-GRF activation. Deletion of the N-terminal PH domain redistributes a large percentage of Ras-GRF from the particulate to the cytosolic fraction of cells and renders the protein insensitive to calcium stimulation. A similar cellular distribution and biological activity are observed when only the core catalytic domain is expressed. Although the PH domain is necessary for particulate association of Ras-GRF, it is not sufficient for targeting the core catalytic domain to this cellular location. This requires the PH domain and the adjacent coiled-coil and IQ sequences. Remarkably, this form of Ras-GRF is constitutively activated. The PH and coiled-coil domains must also perform an additional function, since targeting to the particulate fraction of cells is not sufficient to allow Ras-GRF activation by calcium. A Ras-GRF mutant containing the PH domain from Ras-GTPase-activating protein in place of its own N-terminal PH domain localizes to the particulate fraction of cells but does not respond to calcium. Similar phenotypes are seen with mutant Ras-GRFs containing point mutations in either the PH or coiled-coil domain. These findings argue that the N-terminal PH, coiled-coil, and IQ domains of Ras-GRF function together to connect Ras-GRF to multiple components in the particulate fractions of cells that are required for responsiveness of the protein to calcium signaling.


Assuntos
Proteínas Sanguíneas/química , Cálcio/fisiologia , Fosfoproteínas , Estrutura Terciária de Proteína , Proteínas/química , Células 3T3 , Animais , Linhagem Celular , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Neurônios , Proteínas/genética , Deleção de Sequência , Fatores ras de Troca de Nucleotídeo Guanina , ras-GRF1
16.
Mol Cell Biol ; 19(2): 1334-45, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9891067

RESUMO

The human lbc oncogene product is a guanine nucleotide exchange factor that specifically activates the Rho small GTP binding protein, thus resulting in biologically active, GTP-bound Rho, which in turn mediates actin cytoskeletal reorganization, gene transcription, and entry into the mitotic S phase. In order to elucidate the mechanism of onco-Lbc transformation, here we report that while proto- and onco-lbc cDNAs encode identical N-terminal dbl oncogene homology (DH) and pleckstrin homology (PH) domains, proto-Lbc encodes a novel C terminus absent in the oncoprotein that includes a predicted alpha-helical region homologous to cyto-matrix proteins, followed by a proline-rich region. The lbc proto-oncogene maps to chromosome 15, and onco-lbc represents a fusion of the lbc proto-oncogene N terminus with a short, unrelated C-terminal sequence from chromosome 7. Both onco- and proto-Lbc can promote formation of GTP-bound Rho in vivo. Proto-Lbc transforming activity is much reduced compared to that of onco-Lbc, and a significant increase in transforming activity requires truncation of both the alpha-helical and proline-rich regions in the proto-Lbc C terminus. Deletion of the chromosome 7-derived C terminus of onco-Lbc does not destroy transforming activity, demonstrating that it is loss of the proto-Lbc C terminus, rather than gain of an unrelated C-terminus by onco-Lbc, that confers transforming activity. Mutations of onco-Lbc DH and PH domains demonstrate that both domains are necessary for full transforming activity. The proto-Lbc product localizes to the particulate (membrane) fraction, while the majority of the onco-Lbc product is cytosolic, and mutations of the PH domain do not affect this localization. The proto-Lbc C-terminus alone localizes predominantly to the particulate fraction, indicating that the C terminus may play a major role in the correct subcellular localization of proto-Lbc, thus providing a mechanism for regulating Lbc oncogenic potential.


Assuntos
Proteínas de Ligação ao GTP/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Proteínas de Ancoragem à Quinase A , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Transformação Celular Neoplásica/genética , Quimera/genética , Cromossomos Humanos Par 15/genética , Cromossomos Humanos Par 7/genética , Cricetinae , Primers do DNA/genética , DNA Complementar/genética , Regulação da Expressão Gênica , Rearranjo Gênico , Humanos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Proto-Oncogene Mas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção
17.
Mol Cell Biol ; 20(2): 462-7, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10611224

RESUMO

3Y1 rat fibroblasts overexpressing the epidermal growth factor (EGF) receptor (EGFR cells) become transformed when treated with EGF. A common response to oncogenic and mitogenic stimuli is elevated phospholipase D (PLD) activity. RalA, a small GTPase that functions as a downstream effector molecule of Ras, exists in a complex with PLD1. In the EGFR cells, EGF induced a Ras-dependent activation of RalA. The activation of PLD by EGF in these cells was dependent upon both Ras and RalA. In contrast, EGF-induced activation of Erk1, Erk2, and Jun kinase was dependent on Ras but independent of RalA, indicating divergent pathways activated by EGF and mediated by Ras. The transformed phenotype induced by EGF in the EGFR cells was dependent upon both Ras and RalA. Importantly, overexpression of wild-type RalA or an activated RalA mutant increased PLD activity in the absence of EGF and transformed the EGFR cells. Although overexpression of PLD1 is generally toxic to cells, the EGFR cells not only tolerated PLD1 overexpression but also became transformed in the absence of EGF. These data demonstrate that either RalA or PLD1 can cooperate with EGF receptor to transform cells.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Fosfolipase D/metabolismo , Proteínas ral de Ligação ao GTP , Animais , Divisão Celular , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/genética , Fibroblastos , GTP Fosfo-Hidrolases/genética , Expressão Gênica , Genes Dominantes/genética , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação/genética , Fenótipo , Fosfolipase D/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Oncogene ; 6(12): 2297-304, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1766676

RESUMO

Two inhibitory Ras mutant proteins [(Asn 17) Ras and RAST] were microinjected into NIH3T3 cells in order to compare their inhibitory activity with that of a neutralizing anti-ras antibody. Both mutants were able to block efficiently the mitogenic effects of serum added to quiescent NIH3T3 cells. Furthermore, each of the inhibitors blocked cell cycle progression at the same point as the injected anti-ras antibody, just prior to the initiation of a new round of DNA synthesis. Finally, as with the injected anti-ras antibody, each of the inhibitors was efficiently able to block proliferation and reverse the transformed morphology of cells transformed by tyrosine kinase oncogenes, while cells transformed by serine kinase oncogenes were unaffected. Therefore, results with all three reagents clearly indicate that cellular Ras activity is required in the late G1 phase of the cell cycle and is essential for the maintenance of the transformed phenotype induced by tyrosine but not serine kinase oncogenes. These studies demonstrate the utility of dominant inhibitory mutants as a means of interfering with the activity of cellular oncogenes.


Assuntos
Genes ras , Oncogenes , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Ciclo Celular , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
19.
Oncogene ; 10(10): 1887-93, 1995 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-7761090

RESUMO

Ras-GRF, a guanine-nucleotide exchange factor that activates Ras p21, was tested for its ability to couple to either tyrosine kinase or heterotrimeric G protein signal transduction pathways. Ras-GRF failed to bind the SH2 and SH3 containing adaptor protein Grb2, either in vitro or in vivo. Furthermore, Ras-GRF did not form a stable complex with activated EGF receptor. However, as has been shown previously (Cen et al., 1994), the presence of Ras-GRF in NIH3T3 cells enhanced the activation of Ras induced by serum stimulation. A similar effect was not observed with PDGF stimulation. Moreover, serum stimulation lead to the hyperphosphorylation of Ras-GRF. Both the serum induced super-activation of Ras, and the hyperphosphorylation of Ras-GRF were blocked by pretreatment of cells with the Gi,o inhibitor pertussis toxin, but not by pretreatment with the tyrosine kinase inhibitor genistein. These results suggest that Ras-GRF has the capacity to mediate Ras activation initiated by signals using heterotrimeric G proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Receptores ErbB/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas/metabolismo , Células 3T3 , Animais , Proteínas de Ciclo Celular/metabolismo , Proteína Adaptadora GRB2 , Proteínas Ativadoras de GTPase , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Toxina Pertussis , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteínas/efeitos dos fármacos , Transdução de Sinais , Fatores de Virulência de Bordetella/farmacologia , Proteínas Ativadoras de ras GTPase , Fatores ras de Troca de Nucleotídeo Guanina , ras-GRF1
20.
Oncogene ; 18(33): 4718-25, 1999 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-10467419

RESUMO

Overproduction of urokinase-type plasminogen activator (uPA) and metalloproteases (MMPs) is strongly correlated with tumorigenicity and with invasive and metastatic phenotypes of human and experimental tumors. We demonstrated previously that overproduction of uPA in tumor cells is mediated by a phospholipase D (PLD)- and protein kinase C-dependent mechanism. The oncogenic stimulus of v-Src and v-Ras results in the activation of PLD, which is dependent upon the monomeric GTPase RalA. We have therefore investigated whether RalA plays a role in uPA and MMP overproduction that is observed in response to oncogenic signals. We report here that NIH3T3 cells transformed by both v-Src and v-Ras, constitutively overproduce uPA and that expression of a dominant negative RalA mutant (S28N) blocks overproduction of uPA in both the v-Src-and v-Ras-transformed cells. v-Src and v-Ras also induced an upregulation of the activity of MMP-2 and MMP-9 as detected by zymograms, however only the v-Src induction correlated with MMP protein levels detected by Western blot analysis. The dominant negative RalA mutant blocked increased MMP-2 and 9 overproduction induced by v-Src, but not the increased activity of MMP-2 and 9 induced by v-Ras. And, consistent with a role for the RalA/PLD pathway in mitogenesis and tumor development, the dominant negative RalA mutant completely blocked tumor formation by v-Src- and v-Ras-transformed NIH3T3 cells injected subcutaneously in syngeneic mice. The data presented here implicate RalA and PLD as signaling mediators for tumor formation and protease production by transformed cells.


Assuntos
Transformação Celular Neoplásica/genética , GTP Fosfo-Hidrolases/genética , Genes ras , Genes src , Metaloendopeptidases/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Proteínas ral de Ligação ao GTP , Células 3T3 , Animais , Colagenases/biossíntese , Gelatinases/biossíntese , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA