Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 7(7): 1974-84, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18645007

RESUMO

Neuroblastoma produce angiogenic peptides, and the extent of angiogenesis correlates with tumor progression and poor clinical outcome. Hence, angiogenic factor inhibition represents an important therapeutic option. One of the major drives to tumor angiogenesis is hypoxia, a decrease in oxygen tension that characterizes the tumor microenvironment. We investigated the effects of the topoisomerase I inhibitor, topotecan, on vascular endothelial growth factor (VEGF) induction by hypoxia in advanced-stage human neuroblastoma cells. Topotecan counteracted hypoxic induction of VEGF and decreased angiogenic activity of conditioned medium from hypoxic cultures in vivo in the chick chorioallantoic membrane. Promoter-driven reporter studies showed the role of both hypoxia-inducible factor (HIF)-1alpha and -2alpha in VEGF transcription activation by hypoxia, because (a) overexpression of either protein by cotransfection with expression vectors resulted in VEGF promoter transactivation, which was abrogated by mutation in the HIF-binding site, and (b) targeted knockdown of HIF-1alpha/2alpha by RNA interference inhibited hypoxia-stimulated VEGF transcriptional activity and protein secretion. Topotecan-inhibitory effects on VEGF induction by hypoxia were mediated through suppression of both HIF-1alpha and HIF-2alpha protein accumulation and transactivation properties, which was specific and required ongoing RNA transcription. A similar pattern of results was obtained in cells treated with the hypoxia-mimetic agent, desferrioxamine. These data provide the first evidence that topotecan is a potent inhibitor of HIF-1alpha and HIF-2alpha subunits in hypoxic neuroblastoma cells, leading to decreased VEGF expression and angiogenic activity. An important clinical implication of these findings is that therapies targeted to the HIF pathway have the potential to inhibit neuroblastoma angiogenesis and growth.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Neovascularização Patológica/patologia , Neuroblastoma/irrigação sanguínea , Neuroblastoma/patologia , Topotecan/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Embrião de Galinha , Desferroxamina/farmacologia , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neuroblastoma/genética , Regiões Promotoras Genéticas/genética , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética
2.
Cell Cycle ; 6(16): 2058-70, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17721084

RESUMO

Rho family GTPases play important roles in the regulation of intracellular signals induced by activated heterotrimeric G proteins of the alpha(12/13) family. The alpha(12/13) subunits activate Rho GTPases through direct binding to a group of Rho guanine nucleotide exchange factors (GEFs) characterized by the presence of a G protein signaling-like (RGL) domain. The Rho GEF proto-Dbl, that does not contain a RGL domain, was also found to link Galpha(12/13) signals to Rho. We have explored the effects of activated Galpha(13) and Galpha(13)-associated G protein-coupled receptor (GPCR) agonists on proto-Dbl regulation. We show that activated Galpha(13), but not Galpha(12) or Galpha(q), induces translocation of proto-Dbl to the cell membrane with consequent enlargement of cell body and membrane ruffling. These effects were evident also when Galpha(13)-associated GPCR agonists were used on cells expressing proto-Dbl and were accompanied by the activation of Cdc42 and RhoA GTPases and further downstream effector JNK and p38 kinases. Moreover, we show that both activated Galpha(13) and GPCR agonists stimulate proto-Dbl interaction with ezrin to promote ezrin translocation to the plasma membrane. These results suggest a mechanism by which proto-Dbl and its effector pathways are regulated by Galpha(13)-mediated signals through association with ezrin.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Animais , Transporte Biológico/efeitos dos fármacos , Western Blotting , Bradicinina/farmacologia , Células COS , Chlorocebus aethiops , Proteínas do Citoesqueleto/metabolismo , Imunofluorescência , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Imunoprecipitação , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Células NIH 3T3 , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , RNA Interferente Pequeno/genética , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais/genética , Trombina/farmacologia , Transfecção , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/fisiologia
3.
Cell Cycle ; 5(22): 2657-65, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17172840

RESUMO

Proto-Dbl protein, a guanine nucleotide exchange factor (GEF) for Rho GTPases, is tightly regulated by a combination of mechanisms that involve intra- and intermolecular interaction and N- and C-terminal domain-dependent turnover of the protein. Moreover, the interaction of the PH domain of proto-Dbl with phosphoinositides regulates its subcellular localization and biological activity. Here we show that inhibition of the phosphatidylinositol 3-kinase (PI3K) by molecular and pharmacological inhibitors causes a strong inhibition of proto-Dbl-induced cell proliferation and transformation. Conversely, inhibition of PI3K results in the translocation of proto-Dbl to the plasma membrane, Rac activation and increased membrane ruffles and cell motility. Furthermore, we investigated the signaling molecules involved in proto-Dbl-induced cell transformation and motility and observed that inhibition of PI3K in proto-Dbl expressing cells induces an increase in p38 activity and a decrease in ERK phosphorylation. Our results suggest that proto-Dbl activates distinct downstream effectors to induce morphological changes and cell transformation.


Assuntos
Membrana Celular/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Células COS , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Camundongos , Microscopia de Fluorescência , Morfolinas/farmacologia , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Transfecção
4.
Gene Expr ; 13(3): 155-65, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17193922

RESUMO

The Dbl oncogene is a guanine nucleotide exchange factor for Rho GTPases and its activity has been linked to the regulation of gene transcription. Dbl oncogene expression in NIH3T3 cells leads to changes in morphological and proliferative properties of these cells, inducing a highly transformed phenotype. To gain insights into Dbl oncogene-induced transformation we compared gene expression profiles between Dbl oncogene-transformed and parental NIH3T3 cells by cDNA microarray. We found that Dbl oncogene expression is associated with gene expression modulation involving upregulation of 51 genes and downregulation of 49 genes. Five of the overexpressed genes identified are known to exert antiproliferative functions. Our observations suggest that the expression of Dbl oncogene in NIH3T3 may lead to the induction of genes associated with cell cycle arrest, possibly through the activation of stress-induced kinases.


Assuntos
Expressão Gênica/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Animais , Northern Blotting , Western Blotting , Ciclo Celular , Linhagem Celular Transformada , Células Cultivadas , Ativação Enzimática , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Células NIH 3T3 , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Birth Defects Res A Clin Mol Teratol ; 76(2): 86-95, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16463413

RESUMO

BACKGROUND: The retinoic acid (RA)-catabolizing enzyme Cyp26a1 plays an important role in protecting tailbud tissues from inappropriate exposure to RA. Cyp26a1-null animals exhibit caudal agenesis and spina bifida, imperforate anus, agenesis of the caudal portions of the digestive and urogenital tracts, and malformed lumbosacral skeletal elements. This phenotype closely resembles the most severe form of caudal agenesis in humans. In view of these findings, we investigated a potential involvement of the human CYP26A1 gene in the pathogenesis of caudal regression syndrome (CRS). METHODS: Mutational screening of 49 CRS patients and 132 controls was performed using denaturing high-performance liquid chromatography and sequencing. Differences in the genotype and allele frequency of each SNP were evaluated by chi(2) analysis. The biological significance of the intronic variants was investigated by transfection assays of mutant constructs and by analysis of the splicing patterns with RT-PCR. RESULTS: Mutational screening allowed us to identify 6 SNPs, 4 of which (447 C>G, 1134 G>A, IVS 1+10 G>C, and IVS 4+8 AG>GA) are new. In addition, we describe a novel 2-site haplotype consisting of the 2 intronic SNPs. Both single-locus and haplotype analyses revealed no association with increased risk for CRS. The consequences of the 2 intronic polymorphisms on the mRNA splicing process were also investigated. Moreover, using functional and computational methods we demonstrated that both of these intronic polymorphisms affect the intron splicing efficiency. CONCLUSIONS: Our research did not provide evidence that CYP26A1 has implications for the pathogenesis of human CRS. However, the relationship between CRS risk and the CYP26A1 genotype requires further study with a larger number of genotyped subjects.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Análise Mutacional de DNA , Testes Genéticos , Doenças da Medula Espinal/genética , Medula Espinal/anormalidades , Frequência do Gene , Genótipo , Humanos , Imageamento por Ressonância Magnética , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Ácido Retinoico 4 Hidroxilase , Fatores de Risco , Doenças da Medula Espinal/enzimologia , Doenças da Medula Espinal/fisiopatologia
6.
Cell Cycle ; 4(11): 1675-82, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16294011

RESUMO

The Rho family small GTPase Cdc42 is critical for diverse cellular functions including the regulation of actin organization, cell polarity, intracellular membrane trafficking, transcription, cell cycle progression and cell transformation. Like other members of the Rho family, Cdc42 cycles between the GTP-bound, active state, and the inactive, GDP-bound state under tight regulation, and it is believed that the GTP bound form of Cdc42 represents the active signaling module in eliciting effector activation and cellular responses. The constitutively active mutant, V12, derived from the analogous mutations found in oncogenic Ras that are GTPase-defective, and a "fast-cycling" self-activating mutant, F28, of Cdc42, have been widely in use to study the cellular effects of Cdc42. Here we report that the constitutively active V12 mutant of Cdc42, when stably expressed in cells, could behave in a dominant negative fashion in inhibiting cell proliferation while the F28 mutant was growth stimulatory. The V12 mutant failed to transform NIH3T3 cells while F28 potently stimulated anchorage-independent growth. The growth inhibitory effect of the V12 mutant correlated with activation of JNK2 and suppression of the cyclin D1 and NF-kappaB expressions that were instead upregulated by the F28 mutant. Furthermore, the V12 mutant could suppress, whereas the F28 mutant potentiated or had no effect on, a wide variety of oncogene-induced cell transformation, including that by the Dbl family GEFs Dbl, Vav and Lbc and the oncogenic Ras, ErbB-2, PDGF B or Raf. These results raise the possibility that over-saturation or constitutive activation of Cdc42 signal may negatively impact on cell proliferation and that both the activation and deactivation steps, or the complete GTPase cycle, of Cdc42 is required for proper function.


Assuntos
Linhagem Celular Transformada/citologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/metabolismo , Mutação , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Transformada/metabolismo , Inibidores do Crescimento/fisiologia , Camundongos , Células NIH 3T3 , Proteína Oncogênica p21(ras)/fisiologia , Proteína cdc42 de Ligação ao GTP/fisiologia
7.
Oncogene ; 23(23): 4098-106, 2004 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15064738

RESUMO

Ezrin, a widespread protein involved in cell migration, morphogenesis and cell adhesion, belongs to a large family of proteins known as ERM (ezrin, radixin, moesin). These three closely related proteins are thought to function as linkers between plasma membrane and actin cytoskeleton and their function is regulated by the small GTP-binding protein Rho. It has been previously shown that the active form of radixin can bind in vitro to Dbl, a Rho-specific guanine nucleotide exchange factor, although an in vivo interaction has not yet been demonstrated. In this paper, we attempted to investigate whether ezrin can also associate with Dbl. We show here that Dbl protein can effectively bind both in vitro and in vivo to the N-terminal region (amino acids 1-531) of a constitutively active mutant of ezrin and with the full-length molecule. We found that this binding is mediated by the Dbl pleckstrin homology domain, responsible for the proper subcellular localization of the Dbl protein. Moreover, we show that Dbl induces localization to the plasma membrane of both the active deletion mutant and the full-length ezrin proteins. Finally, we show that the relocalization of ezrin is independent of Dbl GEF activity. These results indicate that Dbl could induce translocation of ezrin to the plasma membrane through a mechanism that does not require ezrin C-terminus phosphorylation by Rho-associated kinases.


Assuntos
Membrana Celular/metabolismo , Fosfoproteínas/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Animais , Células COS , Proteínas do Citoesqueleto , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Células NIH 3T3 , Fosforilação , Transporte Proteico/fisiologia
8.
J Biol Chem ; 277(22): 19745-53, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11907027

RESUMO

The pleckstrin homology (PH) domain of onco-Dbl, a guanine nucleotide exchange factor (GEF) for Cdc42 and RhoA GTPases, interacts with phosphoinositides (PIPs). This interaction modulates both the GEF activity and the targeting to the plasma membrane of onco-Dbl. Conversely, we have previously shown that in proto-Dbl an intramolecular interaction between the N-terminal domain and the PH domain imposes a negative regulation on both the DH and PH functions, suppressing its transforming activity. Here we have further investigated the mode of regulation of proto-Dbl by generating proto-Dbl mutants deleted of the last C-terminal 50 amino acids, which contain a PEST motif, and/or unable to bind to PIPs due to substitutions of the positively charged residues of the PH domain. The PH mutants of proto-Dbl retained a relative weak GEF activity toward Cdc42 and RhoA in vitro, but their RhoA activating potential was impaired in vivo. Further, these mutants lost both the plasma membrane targeting and the transforming activities, contrary to the PH mutants of onco-Dbl that retained the exchange activity both in vitro and in vivo and showed significant, but partially, reduced transforming activity. Deletion of the C-terminal sequences from onco-Dbl did not affect its function, whereas similar deletion of proto-Dbl led to an increase of transforming activity. Analysis of the half-life of the proto-Dbl mutants revealed that deletion of the C-terminal sequences increases the stability of the protein. Overall, the transformation potential of proto-Dbl mutants was associated with an augmented localization of the protein to the plasma membrane and a strong activation of Jun N-terminal kinase activity and transcription of cyclin D1. Together with previous observations, these data suggest that the biological activity of proto-Dbl is tightly regulated by a combination of mechanisms that involve intramolecular interaction, PH binding to PIPs, and the N- and C-terminal domain-dependent turnover of the protein.


Assuntos
Membrana Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas/metabolismo , Células 3T3 , Motivos de Aminoácidos , Animais , Células COS , Domínio Catalítico , Ciclina D1/metabolismo , Fibroblastos/metabolismo , Deleção de Genes , Glutationa Transferase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Luciferases/metabolismo , Camundongos , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , NF-kappa B/metabolismo , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...