Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 13(4): e0194937, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29617434

RESUMO

The E2F transcription factors control key elements of development, including mammary gland branching morphogenesis, with several E2Fs playing essential roles. Additional prior data has demonstrated that loss of individual E2Fs can be compensated by other E2F family members, but this has not been tested in a mammary gland developmental context. Here we have explored the role of the E2Fs and their ability to functionally compensate for each other during mammary gland development. Using gene expression from terminal end buds and chromatin immunoprecipitation data for E2F1, E2F2 and E2F3, we noted both overlapping and unique mammary development genes regulated by each of the E2Fs. Based on our computational findings and the fact that E2Fs share a common binding motif, we hypothesized that E2F transcription factors would compensate for each other during mammary development and function. To test this hypothesis, we generated RNA from E2F1-/-, E2F2-/- and E2F3+/- mouse mammary glands. QRT-PCR on mammary glands during pregnancy demonstrated increases in E2F2 and E2F3a in the E2F1-/- mice and an increase in E2F2 levels in E2F3+/- mice. During lactation we noted that E2F3b transcript levels were increased in the E2F2-/- mice. Given that E2Fs have previously been noted to have the most striking effects on development during puberty, we hypothesized that loss of individual E2Fs would be compensated for at that time. Double mutant mice were generated and compared with the single knockouts. Loss of both E2F1 and E2F2 revealed a more striking phenotype than either knockout alone, indicating that E2F2 was compensating for E2F1 loss. Interestingly, while E2F2 was not able to functionally compensate for E2F3+/- during mammary outgrowth, increased E2F2 expression was observed in E2F3+/- mammary glands during pregnancy day 14.5 and lactation day 5. Together, these findings illustrate the specificity of E2F family members to compensate during development of the mammary gland.


Assuntos
Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F2/metabolismo , Fator de Transcrição E2F3/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Animais , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Feminino , Regulação da Expressão Gênica , Células HCT116 , Humanos , Camundongos , Camundongos Knockout , Gravidez , Interferência de RNA , RNA Interferente Pequeno/metabolismo
2.
Oncotarget ; 6(35): 37316-34, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26512919

RESUMO

The E2F transcriptional activators E2F1, E2F2 and E2F3a regulate many important cellular processes, including DNA replication, apoptosis and centrosome duplication. Previously, we demonstrated that silencing E2F1 or E2F3 suppresses centrosome amplification (CA) and chromosome instability (CIN) in Her2+ breast cancer cells without markedly altering proliferation. However, it is unknown whether and how silencing a single E2F activator, E2F3, affects malignancy of human breast cancer cells. Thus, we injected HCC1954 Her2+ breast cancer cells silenced for E2F3 into mammary fat pads of immunodeficient mice and demonstrated that loss of E2F3 retards tumor growth. Surprisingly, silencing of E2F3 led to significant reductions in mitotic indices relative to vector controls, while the percentage of cells undergoing S phase were not affected. Nek2 is a mitotic kinase commonly upregulated in breast cancers and a critical regulator of Cdk4- or E2F-mediated CA. In this report, we found that Nek2 overexpression rescued back the CA caused by silencing of shE2F3. However, the effects of Nek2 overexpression in affecting tumor growth rates of shE2F3 and shE2F3; GFP cells were inconclusive. Taken together, our results indicate that E2F3 silencing decreases mammary tumor growth by reducing percentage of cells undergoing mitosis.


Assuntos
Neoplasias da Mama/enzimologia , Fator de Transcrição E2F3/deficiência , Inativação Gênica , Mitose , Receptor ErbB-2/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Fator de Transcrição E2F3/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genótipo , Xenoenxertos , Humanos , Camundongos Nus , Quinases Relacionadas a NIMA , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Carga Tumoral
3.
Mol Cell Biol ; 34(17): 3229-43, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24934442

RESUMO

While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.


Assuntos
Fatores de Transcrição E2F/fisiologia , Neoplasias Mamárias Experimentais/etiologia , Animais , Antígenos Transformantes de Poliomavirus , Fatores de Transcrição E2F/deficiência , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/fisiologia , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/fisiologia , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/fisiologia , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Knockout , Células Neoplásicas Circulantes/patologia , Neovascularização Patológica/genética , Infecções por Retroviridae/etiologia , Infecções por Retroviridae/patologia , Transdução de Sinais , Microambiente Tumoral , Infecções Tumorais por Vírus/etiologia , Infecções Tumorais por Vírus/patologia
4.
Cell Death Differ ; 20(7): 931-40, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23558950

RESUMO

The 'activating' E2fs (E2f1-3) are transcription factors that potently induce quiescent cells to divide. Work on cultured fibroblasts suggested they were essential for division, but in vivo analysis in the developing retina and other tissues disproved this notion. The retina, therefore, is an ideal location to assess other in vivo adenovirus E2 promoter binding factor (E2f) functions. It is thought that E2f1 directly induces apoptosis, whereas other activating E2fs only induce death indirectly by upregulating E2f1 expression. Indeed, mouse retinoblastoma (Rb)-null retinal neuron death requires E2f1, but not E2f2 or E2f3. However, we report an entirely distinct mechanism in dying cone photoreceptors. These neurons survive Rb loss, but undergo apoptosis in the cancer-prone retina lacking both Rb and its relative p107. We show that while E2f1 killed Rb/p107 null rod, bipolar and ganglion neurons, E2f2 was required and sufficient for cone death, independent of E2f1 and E2f3. Moreover, whereas E2f1-dependent apoptosis was p53 and p73-independent, E2f2 caused p53-dependent cone death. Our in vivo analysis of cone photoreceptors provides unequivocal proof that E2f-induces apoptosis independent of E2f1, and reveals distinct E2f1- and E2f2-activated death pathways in response to a single tumorigenic insult.


Assuntos
Apoptose/fisiologia , Fator de Transcrição E2F1/fisiologia , Fator de Transcrição E2F2/fisiologia , Fator de Transcrição E2F3/fisiologia , Células Fotorreceptoras Retinianas Cones/patologia , Animais , Apoptose/genética , Divisão Celular/genética , Divisão Celular/fisiologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Retina/patologia , Retina/fisiopatologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/fisiologia , Proteína p107 Retinoblastoma-Like/deficiência , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/fisiologia , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
5.
Dev Biol ; 351(1): 35-45, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21185283

RESUMO

E2F transcription factors regulate the progression of the cell cycle by repression or transactivation of genes that encode cyclins, cyclin dependent kinases, checkpoint regulators, and replication proteins. Although some E2F functions are independent of the Retinoblastoma tumor suppressor (Rb) and related family members, p107 and p130, much of E2F-mediated repression of S phase entry is dependent upon Rb. We previously showed in cultured mouse embryonic fibroblasts that concomitant loss of three E2F activators with overlapping functions (E2F1, E2F2, and E2F3) triggered the p53-p21(Cip1) response and caused cell cycle arrest. Here we report on a dramatic difference in the requirement for E2F during development and in cultured cells by showing that cell cycle entry occurs normally in E2f1-3 triply-deficient epithelial stem cells and progenitors of the developing lens. Sixteen days after birth, however, massive apoptosis in differentiating epithelium leads to a collapse of the entire eye. Prior to this collapse, we find that expression of cell cycle-regulated genes in E2F-deficient lenses is aberrantly high. In a second set of experiments, we demonstrate that E2F3 ablation alone does not cause abnormalities in lens development but rescues phenotypic defects caused by loss of Rb, a binding partner of E2F known to recruit histone deacetylases, SWI/SNF and CtBP-polycomb complexes, methyltransferases, and other co-repressors to gene promoters. Together, these data implicate E2F1-3 in mediating transcriptional repression by Rb during cell cycle exit and point to a critical role for their repressive functions in cell survival.


Assuntos
Proliferação de Células , Fator de Transcrição E2F1/fisiologia , Fator de Transcrição E2F2/fisiologia , Fator de Transcrição E2F3/fisiologia , Proteínas Repressoras/fisiologia , Animais , Apoptose , Sobrevivência Celular , Quebras de DNA de Cadeia Dupla , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F3/deficiência , Células Epiteliais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína do Retinoblastoma/fisiologia , Proteína Supressora de Tumor p53/fisiologia
6.
Nature ; 462(7275): 925-9, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20016601

RESUMO

The activating E2f transcription factors (E2f1, E2f2 and E2f3) induce transcription and are widely viewed as essential positive cell cycle regulators. Indeed, they drive cells out of quiescence, and the 'cancer cell cycle' in Rb1 null cells is E2f-dependent. Absence of activating E2fs in flies or mammalian fibroblasts causes cell cycle arrest, but this block is alleviated by removing repressive E2f or the tumour suppressor p53, respectively. Thus, whether activating E2fs are indispensable for normal division is an area of debate. Activating E2fs are also well known pro-apoptotic factors, providing a defence against oncogenesis, yet E2f1 can limit irradiation-induced apoptosis. In flies this occurs through repression of hid (also called Wrinkled; Smac/Diablo in mammals). However, in mammals the mechanism is unclear because Smac/Diablo is induced, not repressed, by E2f1, and in keratinocytes survival is promoted indirectly through induction of DNA repair targets. Thus, a direct pro-survival function for E2f1-3 and/or its relevance beyond irradiation has not been established. To address E2f1-3 function in normal cells in vivo we focused on the mouse retina, which is a relatively simple central nervous system component that can be manipulated genetically without compromising viability and has provided considerable insight into development and cancer. Here we show that unlike fibroblasts, E2f1-3 null retinal progenitor cells or activated Müller glia can divide. We attribute this effect to functional interchangeability with Mycn. However, loss of activating E2fs caused downregulation of the p53 deacetylase Sirt1, p53 hyperacetylation and elevated apoptosis, establishing a novel E2f-Sirt1-p53 survival axis in vivo. Thus, activating E2fs are not universally required for normal mammalian cell division, but have an unexpected pro-survival role in development.


Assuntos
Apoptose , Fatores de Transcrição E2F/deficiência , Retina/citologia , Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Acetilação , Animais , Divisão Celular , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p21/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Fibroblastos , Camundongos , Camundongos Knockout , Neuroglia/citologia , Neuroglia/metabolismo , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo
7.
Nature ; 462(7275): 930-4, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20016602

RESUMO

In the established model of mammalian cell cycle control, the retinoblastoma protein (Rb) functions to restrict cells from entering S phase by binding and sequestering E2f activators (E2f1, E2f2 and E2f3), which are invariably portrayed as the ultimate effectors of a transcriptional program that commit cells to enter and progress through S phase. Using a panel of tissue-specific cre-transgenic mice and conditional E2f alleles we examined the effects of E2f1, E2f2 and E2f3 triple deficiency in murine embryonic stem cells, embryos and small intestines. We show that in normal dividing progenitor cells E2f1-3 function as transcriptional activators, but contrary to the current view, are dispensable for cell division and instead are necessary for cell survival. In differentiating cells E2f1-3 function in a complex with Rb as repressors to silence E2f targets and facilitate exit from the cell cycle. The inactivation of Rb in differentiating cells resulted in a switch of E2f1-3 from repressors to activators, leading to the superactivation of E2f responsive targets and ectopic cell divisions. Loss of E2f1-3 completely suppressed these phenotypes caused by Rb deficiency. This work contextualizes the activator versus repressor functions of E2f1-3 in vivo, revealing distinct roles in dividing versus differentiating cells and in normal versus cancer-like cell cycles.


Assuntos
Diferenciação Celular , Fatores de Transcrição E2F/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Alelos , Animais , Apoptose , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proliferação de Células , Fatores de Transcrição E2F/deficiência , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/metabolismo
8.
Cell Cycle ; 7(23): 3775-80, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19029823

RESUMO

The E2F transcription factors are key downstream targets of the retinoblastoma protein tumor suppressor. They are known to regulate the expression of genes that control fundamental biological processes including cellular proliferation, apoptosis and differentiation. However, considerable questions remain about the precise roles of the individual E2F family members. This study shows that E2F3 is essential for normal cardiac development. E2F3-loss impairs the proliferative capacity of the embryonic myocardium and most E2f3(-/-) mice die in utero or perinatally with hypoplastic ventricular walls and/or severe atrial and ventricular septal defects. A small fraction of the E2f3(-/-) neonates have hearts that appear grossly normal and they initially survive. However, these animals display ultrastructural defects in the cardiac muscle and ultimately die as a result of congestive heart failure. These data demonstrate a clear role for E2F3 in myocardial and cardiac function during both development and adulthood.


Assuntos
Fator de Transcrição E2F3/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Animais , Proliferação de Células , Fator de Transcrição E2F3/deficiência , Insuficiência Cardíaca/metabolismo , Hipertrofia , Camundongos , Morfogênese , Miocárdio/patologia , Miocárdio/ultraestrutura
9.
Nature ; 454(7208): 1137-41, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18594513

RESUMO

The E2F family is conserved from Caenorhabditis elegans to mammals, with some family members having transcription activation functions and others having repressor functions. Whereas C. elegans and Drosophila melanogaster have a single E2F activator protein and repressor protein, mammals have at least three activator and five repressor proteins. Why such genetic complexity evolved in mammals is not known. To begin to evaluate this genetic complexity, we targeted the inactivation of the entire subset of activators, E2f1, E2f2, E2f3a and E2f3b, singly or in combination in mice. We demonstrate that E2f3a is sufficient to support mouse embryonic and postnatal development. Remarkably, expression of E2f3b or E2f1 from the E2f3a locus (E2f3a(3bki) or E2f3a(1ki), respectively) suppressed all the postnatal phenotypes associated with the inactivation of E2f3a. We conclude that there is significant functional redundancy among activators and that the specific requirement for E2f3a during postnatal development is dictated by regulatory sequences governing its selective spatiotemporal expression and not by its intrinsic protein functions. These findings provide a molecular basis for the observed specificity among E2F activators during development.


Assuntos
Fatores de Transcrição E2F/metabolismo , Desenvolvimento Embrionário , Crescimento , Animais , Células Cultivadas , Fatores de Transcrição E2F/deficiência , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Perda do Embrião/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Deleção de Genes , Genótipo , Crescimento/genética , Camundongos , Camundongos Knockout , Fenótipo
10.
PLoS Biol ; 5(7): e179, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17608565

RESUMO

It has long been known that loss of the retinoblastoma protein (Rb) perturbs neural differentiation, but the underlying mechanism has never been solved. Rb absence impairs cell cycle exit and triggers death of some neurons, so differentiation defects may well be indirect. Indeed, we show that abnormalities in both differentiation and light-evoked electrophysiological responses in Rb-deficient retinal cells are rescued when ectopic division and apoptosis are blocked specifically by deleting E2f transcription factor (E2f) 1. However, comprehensive cell-type analysis of the rescued double-null retina exposed cell-cycle-independent differentiation defects specifically in starburst amacrine cells (SACs), cholinergic interneurons critical in direction selectivity and developmentally important rhythmic bursts. Typically, Rb is thought to block division by repressing E2fs, but to promote differentiation by potentiating tissue-specific factors. Remarkably, however, Rb promotes SAC differentiation by inhibiting E2f3 activity. Two E2f3 isoforms exist, and we find both in the developing retina, although intriguingly they show distinct subcellular distribution. E2f3b is thought to mediate Rb function in quiescent cells. However, in what is to our knowledge the first work to dissect E2f isoform function in vivo we show that Rb promotes SAC differentiation through E2f3a. These data reveal a mechanism through which Rb regulates neural differentiation directly, and, unexpectedly, it involves inhibition of E2f3a, not potentiation of tissue-specific factors.


Assuntos
Células Amácrinas/citologia , Células Amácrinas/metabolismo , Fator de Transcrição E2F3/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Apoptose , Sequência de Bases , Ciclo Celular , Diferenciação Celular/fisiologia , Primers do DNA/genética , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Feminino , Genes do Retinoblastoma , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética
11.
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
12.
Artigo em Inglês | MEDLINE | ID: mdl-16869767

RESUMO

Deregulation of the cell cycle machinery plays a critical role in tumorigenesis. In particular, functional inactivation of the retinoblastoma protein (pRB) is a key event. pRB's tumor suppressive activity is at least partially dependent on its ability to regulate the activity of the E2F transcription factors. E2F controls the expression of genes that encode the cellular proliferation machinery. E2F can also trigger apoptosis when it is inappropriately expressed. Here we present evidence that E2F acts to directly regulate the Arf/p53 tumor surveillance network. In normal cells, a single member of the E2F family, E2F3, participates in the transcriptional silencing of Arf. In response to oncogenic stress, the activating E2Fs, E2F1, 2, and E2F3A, all associate with Arf and promote its transcription. These findings raise the possibility that E2F acts as a sensor of inappropriate versus normal proliferative signals and determines whether or not the Arf/p53 tumor surveillance network is engaged.


Assuntos
Fatores de Transcrição E2F/metabolismo , Genes p53 , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Proteína Supressora de Tumor p14ARF/genética , Animais , Ciclo Celular , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes do Retinoblastoma , Genes Supressores de Tumor , Camundongos , Camundongos Knockout , Proteína do Retinoblastoma/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA