Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Methods Enzymol ; 438: 171-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18413248

RESUMO

Arf GTP-binding proteins and Rho-family GTPases play key roles in regulating membrane remodeling and cytoskeletal reorganization involved in cell movement. Several studies have implicated neurotrophins and their receptors as upstream activators of these small GTP-binding proteins, however, the mechanisms and the cell type specificity of this neurotrophin activity are still under investigation. Here we describe the rationale and protocols used for the dissection of an NT3 activated pathway that leads to the specific activation of Arf6 and Rac1.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Neurônios/metabolismo , Neurotrofina 3/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Fator 6 de Ribosilação do ADP , Animais , Linhagem Celular , Ativação Enzimática , Humanos , Camundongos , Camundongos Mutantes
2.
J Cell Biol ; 173(2): 291-9, 2006 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-16636148

RESUMO

Neurotrophins play an essential role in mammalian development. Most of their functions have been attributed to activation of the kinase-active Trk receptors and the p75 neurotrophin receptor. Truncated Trk receptor isoforms lacking the kinase domain are abundantly expressed during development and in the adult; however, their function and signaling capacity is largely unknown. We show that the neurotrophin-3 (NT3) TrkCT1-truncated receptor binds to the scaffold protein tamalin in a ligand-dependent manner. Moreover, NT3 initiation of this complex leads to activation of the Rac1 GTPase through adenosine diphosphate-ribosylation factor 6 (Arf6). At the cellular level, NT3 binding to TrkCT1-tamalin induces Arf6 translocation to the membrane, which in turn causes membrane ruffling and the formation of cellular protrusions. Thus, our data identify a new signaling pathway elicited by the kinase-deficient TrkCT1 receptor. Moreover, we establish NT3 as an upstream regulator of Arf6.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Receptor trkC/fisiologia , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Fator 6 de Ribosilação do ADP , Linhagem Celular , Células Cultivadas , Humanos , Modelos Biológicos , Isoformas de Proteínas/fisiologia
3.
Blood ; 107(8): 3321-9, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16380453

RESUMO

An important aspect of tumor progression is the ability of cancer cells to escape detection and clearance by the immune system. Recent studies suggest that several tumors express soluble factors interfering with the immune response. Here, we show that semaphorin-3A (Sema-3A), a secreted member of the semaphorin family involved in axonal guidance, organogenesis, and angiogenesis, is highly expressed in several tumor cells. Conditioned media of Sema-3A-transfected COS-7 cells or human recombinant Sema-3A inhibited primary human T-cell proliferation and cytokines production under anti-CD3 plus anti-CD28 stimulating conditions. Sema-3A also inhibited the activation of nonspecific cytotoxic activity in mixed lymphocyte culture (MLC), as measured against K-562 cells. In contrast, suppression of Sema-3A in tumor cells with a small interfering RNA (siRNA) augmented T-cell activation. The inhibitory effect of Sema-3A in T cells is mediated by blockade of Ras/mitogen-activated protein kinase (MAPK) signaling pathway. The presence of Sema-3A increased the activation of the Ras family small GTPase Rap1 and introduction of the dominant-negative mutant of Rap1 (Rap1N17) blunted the immunoinhibitory effects of Sema-3A. These results suggest that Sema-3A inhibits primary T-cell activation and imply that it can contribute to the T-cell dysfunction in the tumor microenvironment.


Assuntos
Ativação Linfocitária/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Neoplasias/imunologia , Semaforina-3A/imunologia , Linfócitos T/imunologia , Substituição de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos , Células COS , Proliferação de Células , Chlorocebus aethiops , Inativação Gênica/imunologia , Células HL-60 , Humanos , Células Jurkat , Ativação Linfocitária/genética , Sistema de Sinalização das MAP Quinases/genética , Neoplasias/genética , Neoplasias/metabolismo , Peptídeos , Mutação Puntual , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Semaforina-3A/biossíntese , Semaforina-3A/genética , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/imunologia
4.
EMBO J ; 24(1): 170-9, 2005 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-15616590

RESUMO

5-Lipoxygenase (5LO) is involved in the production of leukotrienes and reactive oxygen species (ROS) from arachidonic acid. Its strong activation has been associated with several diseases like cancer and neurodegeneration. Here we show that 5LO activity increases during senescence-like growth arrest induced by oncogenic ras or culture history in both human and mouse embryo fibroblasts. Overexpression of 5LO promotes senescence-like growth arrest via a p53/p21-dependent pathway, and this occurs independently of telomerase activity. 5LO stabilizes p53 through phosphorylation at Ser15 and increases expression of the p53-transcriptional target p21. This is achieved by regulating ROS production. Indeed, ROS are increased in 5LO-arrested cells. Antioxidants and a low oxygen environment prevent 5LO-induced growth arrest. Finally, 5LO inhibition reduces the growth arrest induced by oncogenic ras or culture history and these effects are neutralized by the addition of exogenous ROS. These data link the 5LO pathway to oxidative crises of primary fibroblast and suggest that the ability of 5LO to induce senescence-like growth arrest may be important in the pathogenesis of 5LO-associated disorders.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Proliferação de Células , Senescência Celular/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Araquidonato 5-Lipoxigenase/genética , Sobrevivência Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais/fisiologia , Telomerase/metabolismo , Ativação Transcricional
5.
FASEB J ; 18(14): 1740-2, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15375079

RESUMO

5-lipoxygenase (5-LO) promotes cancer cell proliferation and survival by unclear mechanisms. Here, we show that 5-LO expression and activity were induced by genotoxic agents in a p53-independent manner and antagonized p53- or genotoxic drug-induced apoptosis in a variety of cancer cells. 5-LO inhibited p53-governed transactivation of the pro-apoptotic genes bax and pig3 but not of p21(WAF1/CIP1) or mdm2. This may be explained by 5-LO capability to inhibit the binding of p53 to promyelocytic leukemia protein (PML) and p53 subnuclear relocalization into PML-nuclear bodies in response to genotoxic stress. Interestingly, 5-LO activity appears to be involved in nuclear retention and inactivation of wild-type p53 in malignant mesothelioma cells. In these cells, genetic or pharmacological inhibition of 5-LO enabled suppression of in vitro tumorigenicity by low doses of chemotherapeutic drugs. Together, these results uncover novel functions of 5-LO and contribute to the understanding of 5-LO involvement in tumor progression. Moreover, they provide a rationale to the therapeutic use of 5-LO inhibitors to enhance cancer chemosensitivity in selected tumors.


Assuntos
Apoptose , Araquidonato 5-Lipoxigenase/metabolismo , Núcleo Celular/metabolismo , Mutagênicos/toxicidade , Neoplasias/enzimologia , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Estruturas do Núcleo Celular/química , Resistencia a Medicamentos Antineoplásicos , Humanos , Proteínas de Neoplasias/análise , Neoplasias/química , Neoplasias/metabolismo , Proteínas Nucleares/análise , Proteína da Leucemia Promielocítica , Fatores de Transcrição/análise , Transcrição Gênica , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteínas Supressoras de Tumor
6.
J Biol Chem ; 279(45): 46706-14, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15337769

RESUMO

Malignant mesothelioma (MM) is strongly resistant to conventional chemotherapy by unclear mechanisms. We and others have previously reported that cytokine- and growth factor-mediated signal transduction is involved in the growth and progression of MM. Here, we identified a pathway that involves stem cell factor (SCF)/c-Kit/Slug in mediating multidrug resistance of MM cells. When we compared gene expression profiles between five MM cells and their multidrug-resistant (MM DX) sublines, we found that MM DX cells expressed both SCF and c-Kit and had higher mRNA levels of Slug. Knockdown of c-Kit or Slug expression with their respective small interfering RNA sensitized MM DX cells to the induction of apoptosis by different chemotherapeutic agents, including doxorubicin, paclitaxel, and vincristine. Transfection of c-Kit in parental MM cells in the presence of SCF up-regulated Slug and increased resistance to the chemotherapeutic agents. Moreover, MM cells expressing Slug showed a similar increased resistance to the chemotherapeutic agents. These results indicate that induction of Slug by autocrine production of SCF and c-Kit activation plays a key role in conferring a broad spectrum chemoresistance on MM cells and reveal a novel signal transduction pathway for pharmacological or genetic intervention of MM patients.


Assuntos
Resistência a Múltiplos Medicamentos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Mesotelioma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Apoptose , Linhagem Celular Tumoral , Separação Celular , Meios de Cultivo Condicionados/farmacologia , Citocinas/biossíntese , DNA Complementar/metabolismo , Progressão da Doença , Relação Dose-Resposta a Droga , Regulação para Baixo , Doxorrubicina/farmacologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Imunoprecipitação , Mesotelioma/patologia , Paclitaxel/farmacologia , Fenótipo , Plasmídeos/metabolismo , Proteínas Tirosina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Transfecção , Regulação para Cima , Verapamil/farmacologia , Vincristina/farmacologia
7.
FASEB J ; 18(2): 358-60, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14656993

RESUMO

Vascular endothelial growth factor (VEGF) and semaphorin-3A (Sema-3A) play important roles in the transduction of promitotic and antimitotic signals, respectively. Here, we report that these conflicting signals are integrated via negative feedback between VEGF and Sema-3A pathways in several primary normal, but not malignant, mesothelial cells. Unlike malignant mesothelial (MM) cells, in which VEGF induces cell proliferation, normal mesothelial (NM) cell growth was repressed by VEGF. Although both cell-types expressed an overlapping set of VEGF tyrosine-kinase receptors, only in NM cells VEGF exposure entails a p38 mitogen-activated protein kinase (MAPK)-dependent increased of Sema-3A production. Inhibition of p38 MAPK (by SB202190 and SB203580) or a dominant-negative mutant of Sema-3A receptor plexin-A1 reversed the inhibitory effects of VEGF in NM cells, increasing cyclin D1 synthesis and cell growth. Conversely, sustained activation of p38 MAPK by the p38 MAPK-activating kinases MKK3 and MKK6 or transfection with Sema-3A inhibited VEGF-induced cyclin D1 up-regulation and MM cell proliferation. Therefore, these results delineate a new role of Sema-3A in VEGF function mediated by p38 MAPK and suggest that the abrogation of regulated Sema-3A expression is responsible for VEGF-driven growth of tumor cells.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/patologia , Semaforina-3A/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Ciclina D1/biossíntese , Ciclina D1/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Retroalimentação Fisiológica , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Semaforina-3A/genética , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA