Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 321(1): 64-76, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18582454

RESUMO

The FGF signaling pathway plays essential roles in endochondral ossification by regulating osteoblast proliferation and differentiation, chondrocyte proliferation, hypertrophy, and apoptosis. FGF signaling is controlled by the complementary action of both positive and negative regulators of the signal transduction pathway. The Spry proteins are crucial regulators of receptor tyrosine kinase-mediated MAPK signaling activity. Sprys are expressed in close proximity to FGF signaling centers and regulate FGFR-ERK-mediated organogenesis. During endochondral ossification, Spry genes are expressed in prehypertrophic and hypertrophic chondrocytes. Using a conditional transgenic approach in chondrocytes in vivo, the forced expression of Spry1 resulted in neonatal lethality with accompanying skeletal abnormalities resembling thanatophoric dysplasia II, including increased apoptosis and decreased chondrocyte proliferation in the presumptive reserve and proliferating zones. In vitro chondrocyte cultures recapitulated the inhibitory effect of Spry1 on chondrocyte proliferation. In addition, overexpression of Spry1 resulted in sustained ERK activation and increased expression of p21 and STAT1. Immunoprecipitation experiments revealed that Spry1 expression in chondrocyte cultures resulted in decreased FGFR2 ubiquitination and increased FGFR2 stability. These results suggest that constitutive expression of Spry1 in chondrocytes results in attenuated FGFR2 degradation, sustained ERK activation, and up-regulation of p21Cip and STAT1 causing dysregulated chondrocyte proliferation and terminal differentiation.


Assuntos
Condrócitos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Diferenciação Celular , Proliferação de Células , Condrócitos/citologia , Camundongos , Camundongos Transgênicos , Osteogênese , Fator de Transcrição STAT1/metabolismo , Ubiquitinação , Regulação para Cima
2.
Cell Signal ; 18(11): 1958-66, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16603339

RESUMO

Sef (similar expression to fgf genes) is a member of the fibroblast growth factor (FGF) synexpression group that negatively regulates FGF receptor (FGFR) signaling in zebrafish during early embryonic development and in mammalian cell culture systems. The mechanism by which Sef exerts its inhibitory effect remains controversial. It has been reported that Sef functions either through binding to and inhibiting FGFR1 activation or by acting downstream of FGF receptors at the level of MEK/ERK kinases. In both cases, the intracellular domain of Sef was found to play a role in the inhibitory function of Sef. Here we demonstrated that both extracellular and transmembrane domains of Sef contributed to Sef-mediated negative regulation of FGF signaling. In fact, over-expression studies in NIH3T3 cells showed that a truncated mutant of Sef, which lacks the intracellular domain (SefECTM), exerted the inhibitory activity on FGF signaling by inhibiting FGFR1 tyrosine phosphorylation and subsequent activation of the Raf/MEK/ERK signaling cascade. We also showed that SefECTM associated with FGFR1, and inhibited FGF-induced ERK activation in HEK293T cells. Furthermore, we demonstrated that the over-expression of SefECTM was able to inhibit the function of a constitutively activated form of FGFR1, FGFR1-C289R, but not FGFR1-K562E. Finally, we found that SefECTM reduced cell viability when over-expressed in human umbilical vein endothelial cells (HUVEC). These data provide additional insight into the structure-activity relationship in the mechanism of inhibitory action of Sef on FGFR1-mediated signaling.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas de Membrana/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Células NIH 3T3 , Fragmentos de Peptídeos/farmacologia , Fosforilação , Estrutura Terciária de Proteína , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica
3.
J Biol Chem ; 279(37): 38099-102, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15277532

RESUMO

Sef was recently identified as a negative regulator of fibroblast growth factor (FGF) signaling in a genetic screen of zebrafish and subsequently in mouse and humans. By inhibiting FGFR1 tyrosine phosphorylation and/or Ras downstream events, Sef inhibits FGF-mediated ERK activation and cell proliferation as well as PC12 cell differentiation. Here we show that Sef and a deletion mutant of Sef lacking the extracellular domain (SefIC) physically interact with TAK1 (transforming growth factor-beta-associated kinase) and activate JNK through a TAK1-MKK4-JNK pathway. Sef and SefIC overexpression also resulted in apoptotic cell death, while dominant negative forms of MKK4 and TAK1 blocked Sef-mediated JNK activation and attendant 293T cell apoptosis. These investigations reveal a novel activating function of Sef that is distinct from its inhibitory effect on FGF receptor signaling and ERK activation.


Assuntos
Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase Quinases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Receptores de Interleucina/metabolismo , Animais , Diferenciação Celular , Divisão Celular , Linhagem Celular , Relação Dose-Resposta a Droga , Ativação Enzimática , Citometria de Fluxo , Deleção de Genes , Genes Dominantes , Humanos , Immunoblotting , MAP Quinase Quinase 4 , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Células PC12 , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Ratos , Transdução de Sinais , Fatores de Tempo , Transfecção , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...