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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
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
2.
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
3.
Mol Cell Biol ; 27(6): 2283-93, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17210634

RESUMO

The tumor suppressor function of the retinoblastoma protein pRB is largely dependent upon its capacity to inhibit the E2F transcription factors and thereby cell proliferation. Attempts to study the interplay between pRB and the E2Fs have been hampered by the prenatal death of Rb; E2f nullizygous mice. In this study, we isolated Rb; E2f3 mutant embryonic stem cells and generated Rb(-/-); E2f3(-/-) chimeric mice, thus bypassing the lethality of the Rb(-/-); E2f3(-/-) germ line mutant mice. We show that loss of E2F3 has opposing effects on two of the known developmental defects arising in Rb(-/-) chimeras; it suppresses the formation of cataracts while aggravating the retinal dysplasia. This model system also allows us to assess how E2f3 status influences tumor formation in Rb(-/-) tissues. We find that E2f3 is dispensable for the development of pRB-deficient pituitary and thyroid tumors. In contrast, E2f3 inactivation completely suppresses the pulmonary neuroendocrine hyperplasia arising in Rb(-/-) chimeric mice. This hyperproliferative state is thought to represent the preneoplastic lesion of small-cell lung carcinoma. Therefore, our observation highlights a potential role for E2F3 in the early stages of this tumor type.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Fator de Transcrição E2F3/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteína do Retinoblastoma/metabolismo , Animais , Separação Celular , Transformação Celular Neoplásica/genética , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Células-Tronco Embrionárias/metabolismo , Olho/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Mutação/genética , Neoplasias/genética , Especificidade de Órgãos , Doenças da Hipófise/genética , Doenças da Hipófise/metabolismo , Doenças da Hipófise/patologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia
4.
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
5.
Mol Cell Biol ; 22(8): 2663-72, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11909960

RESUMO

The E2F1, -2, and -3 transcription factors are key downstream targets of the retinoblastoma protein (pRB) tumor suppressor that drive expression of proliferation-associated genes. Here we use mutant mouse strains to investigate E2F3's role in vivo. We show that E2F3 is essential for embryonic viability in the pure 129/Sv background but the presence of C57BL/6 alleles yields some adult survivors. Although growth retarded, surviving E2f3(-/-) animals are initially healthy. However, they die prematurely, exhibiting no obvious tumor phenotype but with the typical signs of congestive heart failure. The defects are completely distinct from those arising in E2f1 mutant mice (S. J. Field et al., Cell 85:549-561; 1996; L. Yamasaki et al., Cell 85:537-548, 1996), supporting the prevailing view that these E2Fs must have some unique biological functions in vivo. To test this model, we examined the phenotypes of E2f1 E2f3 compound mutant mice. Almost all of the developmental and age-related defects arising in the individual E2f1 or E2f3 mice were exacerbated by the mutation of the other E2f. Thus, E2F1 and E2F3 appear to play critical, overlapping roles in the development and maintenance of a variety of tissues. Importantly, this study did identify one major difference in the properties of E2F1 and E2F3: either alone or in combination with E2F1 loss, E2f3 mutation did not increase the incidence of tumor formation. These data strongly suggest that tumor suppression is a specific property of E2F1 and not E2F3.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F3 , Desenvolvimento Embrionário e Fetal/genética , Feminino , Insuficiência Cardíaca/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Modelos Cardiovasculares , Neoplasias Experimentais/genética , Fenótipo , Fatores de Transcrição/deficiência
6.
Dev Biol ; 305(2): 564-76, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17383628

RESUMO

The airway epithelium is comprised of specialized cell types that play key roles in protecting the lungs from environmental insults. The cellular composition of the murine respiratory epithelium is established during development and different cell types populate specific regions along the airway. Here we show that E2f4-deficiency leads to an absence of ciliated cells from the entire airway epithelium and the epithelium of the submucosal glands in the paranasal sinuses. This defect is particularly striking in the nasal epithelium of E2f4-/- mice where ciliated cells are replaced by columnar secretory cells that produce mucin-like substances. In addition, in the proximal lung, E2f4 loss causes a reduction in Clara cell marker expression indicating that Clara cell development is also affected. These defects arise during embryogenesis and, in the nasal epithelium, appear to be independent of any changes in cell proliferation, the principal process regulated by members of the E2f family of transcription factors. We therefore conclude that E2f4 is required to determine the appropriate development of the airway epithelium. Importantly, the combination of no ciliated cells and excess mucous cells can account for the chronic rhinitis and increased susceptibility to opportunistic infections that causes the postnatal lethality of E2f4 mutant mice.


Assuntos
Fator de Transcrição E2F4/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mucosa Respiratória/embriologia , Animais , Proliferação de Células , Cílios/patologia , Cílios/ultraestrutura , Fator de Transcrição E2F4/deficiência , Fator de Transcrição E2F4/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Respiratória/patologia , Mucosa Respiratória/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA