Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 466(7310): 1110-4, 2010 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-20686481

RESUMO

Mutation of the retinoblastoma gene (RB1) tumour suppressor occurs in one-third of all human tumours and is particularly associated with retinoblastoma and osteosarcoma. Numerous functions have been ascribed to the product of the human RB1 gene, the retinoblastoma protein (pRb). The best known is pRb's ability to promote cell-cycle exit through inhibition of the E2F transcription factors and the transcriptional repression of genes encoding cell-cycle regulators. In addition, pRb has been shown in vitro to regulate several transcription factors that are master differentiation inducers. Depending on the differentiation factor and cellular context, pRb can either suppress or promote their transcriptional activity. For example, pRb binds to Runx2 and potentiates its ability to promote osteogenic differentiation in vitro. In contrast, pRb acts with E2F to suppress peroxisome proliferator-activated receptor gamma subunit (PPAR-gamma), the master activator of adipogenesis. Because osteoblasts and adipocytes can both arise from mesenchymal stem cells, these observations suggest that pRb might play a role in the choice between these two fates. However, so far, there is no evidence for this in vivo. Here we use mouse models to address this hypothesis in mesenchymal tissue development and tumorigenesis. Our data show that Rb status plays a key role in establishing fate choice between bone and brown adipose tissue in vivo.


Assuntos
Tecido Adiposo Marrom/citologia , Diferenciação Celular , Linhagem da Célula , Osteoblastos/citologia , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lipoma/fisiopatologia , Camundongos , Mutação/genética , PPAR gama/metabolismo , Sarcoma/fisiopatologia
2.
Cell Cycle ; 9(13): 2620-8, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20581455

RESUMO

The E2F family of transcription factors, in association with pocket protein family members, are important for regulating genes required for cellular proliferation. The most abundant E2F, E2F4, is implicated in maintaining the G(0)/G(1) cell cycle state via transcriptional repression of genes that encode proteins required for S-phase progression. Here, we investigate E2F4's role in bone development using E2f4 germline mutant mice. We find that mutation of E2f4 impairs the formation of several bones that arise through intramembranous or endochondral ossification. The most severe defect occurred in the calvarial bones of the skull where we observed a striking delay in their ossification. In vivo and in vitro analyses established that E2F4 loss did not block the intrinsic differentiation potential of calvarial osteoblast progenitors. However, our data showed that E2f4 mutation elevated proliferation in the developing calvaria in vivo and it increased the endogenous pool of undifferentiated progenitor cells. These data suggest that E2F4 plays an important role in enabling osteoblast progenitors to exit the cell cycle and subsequently differentiate thereby contributing to the commitment of these cells to the bone lineage.


Assuntos
Fator de Transcrição E2F4/genética , Embrião de Mamíferos/patologia , Mutação/genética , Osteogênese , Crânio/embriologia , Crânio/fisiopatologia , Células-Tronco/patologia , Fosfatase Alcalina/metabolismo , Animais , Matriz Óssea/metabolismo , Cartilagem/embriologia , Cartilagem/patologia , Proliferação de Células , Fator de Transcrição E2F4/deficiência , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/fisiopatologia , Camundongos , Camundongos Mutantes , Osteoblastos/enzimologia , Osteoblastos/patologia , Crânio/patologia , Células-Tronco/metabolismo
3.
Cell Cycle ; 9(2): 371-6, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20023434

RESUMO

The retinoblastoma tumor suppressor protein pRB functions, at least in part, by directly binding to and modulating the activity of the E2F transcription factors. Previous studies have shown that both E2F4 and pRB play important roles in fetal erythropoiesis. Given that these two proteins interact directly we investigated the overlap of E2F4 and pRB function in this process by analyzing E2f4(-/-), conditional Rb knockout (Rb(1lox/1lox)), and compound E2f4(-/-);Rb(1lox/1lox) embryos. At E15.5 E2f4(-/-) and Rb(1lox/1lox) fetal erythroid cells display distinct abnormalities in their differentiation profiles. When cultured in vitro, both E2f4(-/-) and Rb(1lox/1lox) erythroid cells show defects in cell cycle progression. Surprisingly, analysis of cell cycle profiling suggests that E2F4 and pRB control cell cycle exit through different mechanisms. Moreover, only pRB, but not E2F4, promotes cell survival in erythroid cells. We observed an additive rather than a synergistic impact upon the erythroid defects in the compound E2f4(-/-);Rb(1lox/1lox) embryos. We further found that fetal liver macrophage development is largely normal regardless of genotype. Taken together, our results show that E2F4 and pRB play independent cell-intrinsic roles in fetal erythropoiesis.


Assuntos
Fator de Transcrição E2F4/metabolismo , Células Eritroides/citologia , Eritropoese , Proteína do Retinoblastoma/metabolismo , Animais , Apoptose , Diferenciação Celular , Sobrevivência Celular , Fator de Transcrição E2F4/genética , Fator de Transcrição E2F4/fisiologia , Embrião de Mamíferos/citologia , Feto , Técnicas de Inativação de Genes , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/fisiologia
4.
Proc Natl Acad Sci U S A ; 106(35): 14932-6, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19706423

RESUMO

The retinoblastoma tumor-suppressor protein, pRb, is a member of the pocket protein family that includes p107 and p130. These proteins have well-defined roles in regulating entry into and exit from the cell cycle and also have cell cycle-independent roles in facilitating differentiation. Here we investigate the overlap between pocket protein's function during embryonic development by using conditional mutant alleles to generate Rb;p107 double-mutant embryos (DKOs) that develop in the absence of placental defects. These DKOs die between e13.5 and e14.5, much earlier than either the conditional Rb or the germline p107 single mutants, which survive to birth or are largely viable, respectively. Analyses of the e13.5 DKOs shows that p107 mutation exacerbates the phenotypes resulting from pRb loss in the central nervous system and lens, but not in the peripheral nervous system. In addition, these embryos exhibit novel phenotypes, including increased proliferation of blood vessel endothelial cells, and heart defects, including double-outlet right ventricle (DORV). The DORV is caused, at least in part, by a defect in blood vessel endothelial cells and/or heart mesenchymal cells. These findings demonstrate novel, overlapping functions for pRb and p107 in numerous murine tissues.


Assuntos
Embrião de Mamíferos/metabolismo , Cardiopatias/metabolismo , Mutação , Proteína do Retinoblastoma/deficiência , Proteína p107 Retinoblastoma-Like/genética , Animais , Apoptose , Proliferação de Células , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Embrião de Mamíferos/irrigação sanguínea , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias/embriologia , Cardiopatias/genética , Cardiopatias/patologia , Cristalino/citologia , Cristalino/metabolismo , Masculino , Camundongos , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like/deficiência
5.
Mol Cancer Res ; 6(9): 1440-51, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18819932

RESUMO

Mutation of the retinoblastoma (RB) tumor suppressor gene is strongly linked to osteosarcoma formation. This observation and the documented interaction between the retinoblastoma protein (pRb) and Runx2 suggests that pRb is important in bone development. To assess this hypothesis, we used a conditional knockout strategy to generate pRb-deficient embryos that survive to birth. Analysis of these embryos shows that Rb inactivation causes the abnormal development and impaired ossification of several bones, correlating with an impairment in osteoblast differentiation. We further show that Rb inactivation acts to promote osteoblast differentiation in vitro and, through conditional analysis, establish that this occurs in a cell-intrinsic manner. Although these in vivo and in vitro differentiation phenotypes seem paradoxical, we find that Rb-deficient osteoblasts have an impaired ability to exit the cell cycle both in vivo and in vitro that can explain the observed differentiation defects. Consistent with this observation, we show that the cell cycle and the bone defects in Rb-deficient embryos can be suppressed by deletion of E2f1, a known proliferation inducer that acts downstream of Rb. Thus, we conclude that pRb plays a key role in regulating osteoblast differentiation by mediating the inhibition of E2F and consequently promoting cell cycle exit.


Assuntos
Desenvolvimento Ósseo/fisiologia , Doenças Ósseas/patologia , Diferenciação Celular , Osteoblastos/citologia , Osteogênese/fisiologia , Proteína do Retinoblastoma/fisiologia , Animais , Ciclo Celular , Proliferação de Células , Fator de Transcrição E2F1/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Integrases/metabolismo , Camundongos , Camundongos Knockout , Osteoblastos/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Proc Natl Acad Sci U S A ; 105(33): 11851-6, 2008 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-18697945

RESUMO

Mutation of the RB-1 and p53 tumor suppressors is associated with the development of human osteosarcoma. With the goal of generating a mouse model of this disease, we used conditional and transgenic mouse strains to inactivate Rb and/or p53 specifically in osteoblast precursors. The resulting Rb;p53 double mutant (DKO) animals are viable but develop early onset osteosarcomas with complete penetrance. These tumors display many of the characteristics of human osteosarcomas, including being highly metastatic. We established cell lines from the DKO osteosarcomas to further investigate their properties. These immortalized cell lines are highly proliferative and they retain their tumorigenic potential, as judged by their ability to form metastatic tumors in immunocompromised mice. Moreover, they can be induced to differentiate and, depending on the inductive signal, will adopt either the osteogenic or adipogenic fate. Consistent with this multipotency, a significant portion of these tumor cells express Sca-1, a marker that is typically associated with stem cells/uncommitted progenitors. By assaying sorted cells in transplant assays, we demonstrate that the tumorigenicity of the osteosarcoma cell lines correlates with the presence of the Sca-1 marker. Finally, we show that loss of Rb and p53 in Sca-1-positive mesenchymal stem/progenitor cells is sufficient to yield transformed cells that can initiate osteosarcoma formation in vivo.


Assuntos
Linhagem da Célula , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antígenos Ly/metabolismo , Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Genótipo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Camundongos Transgênicos , Mutação/genética , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Transplante de Neoplasias , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Osteossarcoma/genética , Proteína do Retinoblastoma/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA