Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Cell ; 6(4): 347-60, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15488758

RESUMO

We identify a novel alternative TrkA splice variant, TrkAIII, with deletion of exons 6, 7, and 9 and functional extracellular IG-C1 and N-glycosylation domains, that exhibits expression restricted to undifferentiated early neural progenitors, human neuroblastomas (NBs), and a subset of other neural crest-derived tumors. This NGF-unresponsive isoform is oncogenic in NIH3T3 cells and promotes tumorigenic NB cell behavior in vitro and in vivo (cell survival, xenograft growth, angiogenesis) resulting from spontaneous tyrosine kinase activity and IP3K/Akt/NF-kappaB but not Ras/MAPK signaling. TrkAIII antagonizes NGF/TrkAI signaling, which is responsible for NB growth arrest and differentiation through Ras/MAPK, and its expression is promoted by hypoxia at the expense of NGF-responsive receptors, providing a mechanism for converting NGF/TrkA/Ras/MAPK antioncogenic signals to TrkAIII/IP3K/Akt/NF-kappaB tumor-promoting signals during tumor progression.


Assuntos
Processamento Alternativo/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Receptor trkA/genética , Receptor trkA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia/genética , Camundongos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Neovascularização Patológica , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Neuroblastoma/irrigação sanguínea , Neuroblastoma/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Ligação Proteica , Receptor trkA/antagonistas & inibidores , Receptor trkA/química , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Fosfolipases Tipo C/metabolismo
2.
J Cell Biol ; 158(4): 731-40, 2002 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12186855

RESUMO

Expansion and fate choice of pluripotent stem cells along the neuroectodermal lineage is regulated by a number of signals, including EGF, retinoic acid, and NGF, which also control the proliferation and differentiation of central nervous system (CNS) and peripheral nervous system (PNS) neural progenitor cells. We report here the identification of a novel gene, REN, upregulated by neurogenic signals (retinoic acid, EGF, and NGF) in pluripotent embryonal stem (ES) cells and neural progenitor cell lines in association with neurotypic differentiation. Consistent with a role in neural promotion, REN overexpression induced neuronal differentiation as well as growth arrest and p27Kip1 expression in CNS and PNS neural progenitor cell lines, and its inhibition impaired retinoic acid induction of neurogenin-1 and NeuroD expression. REN expression is developmentally regulated, initially detected in the neural fold epithelium of the mouse embryo during gastrulation, and subsequently throughout the ventral neural tube, the outer layer of the ventricular encephalic neuroepithelium and in neural crest derivatives including dorsal root ganglia. We propose that REN represents a novel component of the neurogenic signaling cascade induced by retinoic acid, EGF, and NGF, and is both a marker and a regulator of neuronal differentiation.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Fatores de Transcrição , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Biomarcadores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Células Cultivadas , Clonagem Molecular , Inibidor de Quinase Dependente de Ciclina p27 , DNA Complementar , Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Dados de Sequência Molecular , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/metabolismo , Transferases , Tretinoína/farmacologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
3.
J Neurosci ; 25(36): 8338-46, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16148242

RESUMO

During the early development of the cerebellum, a burst of granule cell progenitor (GCP) proliferation occurs in the outer external granule layer (EGL), which is sustained mainly by Purkinje cell-derived Sonic Hedgehog (Shh). Shh response is interrupted once GCPs move into the inner EGL, where granule progenitors withdraw proliferation and start differentiating and migrating toward the internal granule layer (IGL). Failure to interrupt Shh signals results in uncoordinated proliferation and differentiation of GCPs and eventually leads to malignancy (i.e., medulloblastoma). The Shh inhibitory mechanisms that are responsible for GCP growth arrest and differentiation remain unclear. Here we report that REN, a putative tumor suppressor frequently deleted in human medulloblastoma, is expressed to a higher extent in nonproliferating inner EGL and IGL granule cells than in highly proliferating outer EGL cells. Accordingly, upregulated REN expression occurs along GCP differentiation in vitro, and, in turn, REN overexpression promotes growth arrest and increases the proportion of p27/Kip1+ GCPs. REN also impairs both Gli2-dependent gene transcription and Shh-enhanced expression of the target Gli1 mRNA, thus antagonizing the Shh-induced effects on the proliferation and differentiation of cultured GCPs. Conversely, REN functional knock-down impairs Hedgehog antagonism and differentiation and sustains the proliferation of GCPs. Finally, REN enhances caspase-3 activation and terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling apoptotic GCP numbers; therefore, the pattern of REN expression, its activity, and its antagonism on the Hedgehog pathway suggest that this gene may represent a restraint of Shh signaling at the outer to inner EGL GCP transitions. Medulloblastoma-associated REN loss of function might withdraw such a limiting signal for immature cell expansion, thus favoring tumorigenesis.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Cerebelo/fisiologia , Bulbo/fisiologia , Proteínas do Tecido Nervoso/genética , Transativadores/antagonistas & inibidores , Animais , Caspase 3 , Caspases/metabolismo , Técnicas de Cultura de Células , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/genética , Ativação Enzimática , Regulação da Expressão Gênica/fisiologia , Proteínas Hedgehog , Camundongos , Proteínas Recombinantes/metabolismo , Transferases
4.
Proc Natl Acad Sci U S A ; 101(29): 10833-8, 2004 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-15249678

RESUMO

Hedgehog signaling is suggested to be a major oncogenic pathway in medulloblastoma, which arises from aberrant development of cerebellar granule progenitors. Allelic loss of chromosome 17p has also been described as the most frequent genetic defect in this human neoplasia. This observation raises the question of a possible interplay between 17p deletion and the Hedgehog tumorigenic pathway. Here, we identify the human orthologue of mouse REN(KCTD11), previously reported to be expressed in differentiating and low proliferating neuroblasts. Human REN(KCTD11) maps to 17p13.2 and displays allelic deletion as well as significantly reduced expression in medulloblastoma. REN(KCTD11) inhibits medulloblastoma cell proliferation and colony formation in vitro and suppresses xenograft tumor growth in vivo. REN(KCTD11) seems to inhibit medulloblastoma growth by negatively regulating the Hedgehog pathway because it antagonizes the Gli-mediated transactivation of Hedgehog target genes, by affecting Gli1 nuclear transfer, and its growth inhibitory activity is impaired by Gli1 inactivation. Therefore, we identify REN(KCTD11) as a suppressor of Hedgehog signaling and suggest that its inactivation might lead to a deregulation of the tumor-promoting Hedgehog pathway in medulloblastoma.


Assuntos
Neoplasias Cerebelares/genética , Meduloblastoma/genética , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Alelos , Animais , Proteínas de Ciclo Celular , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Cromossomos Humanos Par 17 , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Canais de Potássio/genética , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transferases , Transplante Heterólogo , Proteína GLI1 em Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa