Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 29(8): 1596-1610, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35322202

RESUMO

Multiciliated cells (MCCs) in the brain reside in the ependyma and the choroid plexus (CP) epithelia. The CP secretes cerebrospinal fluid that circulates within the ventricular system, driven by ependymal cilia movement. Tumors of the CP are rare primary brain neoplasms mostly found in children. CP tumors exist in three forms: CP papilloma (CPP), atypical CPP, and CP carcinoma (CPC). Though CPP and atypical CPP are generally benign and can be resolved by surgery, CPC is a particularly aggressive and little understood cancer with a poor survival rate and a tendency for recurrence and metastasis. In contrast to MCCs in the CP epithelia, CPCs in humans are characterized by solitary cilia, frequent TP53 mutations, and disturbances to multiciliogenesis program directed by the GMNC-MCIDAS transcriptional network. GMNC and MCIDAS are early transcriptional regulators of MCC fate differentiation in diverse tissues. Consistently, components of the GMNC-MCIDAS transcriptional program are expressed during CP development and required for multiciliation in the CP, while CPC driven by deletion of Trp53 and Rb1 in mice exhibits multiciliation defects consequent to deficiencies in the GMNC-MCIDAS program. Previous studies revealed that abnormal NOTCH pathway activation leads to CPP. Here we show that combined defects in NOTCH and Sonic Hedgehog signaling in mice generates tumors that are similar to CPC in humans. NOTCH-driven CP tumors are monociliated, and disruption of the NOTCH complex restores multiciliation and decreases tumor growth. NOTCH suppresses multiciliation in tumor cells by inhibiting the expression of GMNC and MCIDAS, while Gmnc-Mcidas overexpression rescues multiciliation defects and suppresses tumor cell proliferation. Taken together, these findings indicate that reactivation of the GMNC-MCIDAS multiciliogenesis program is critical for inhibiting tumorigenesis in the CP, and it may have therapeutic implications for the treatment of CPC.


Assuntos
Carcinoma , Proteínas de Ciclo Celular , Neoplasias do Plexo Corióideo , Proteínas Nucleares , Animais , Carcinoma/genética , Proteínas de Ciclo Celular/genética , Neoplasias do Plexo Corióideo/genética , Neoplasias do Plexo Corióideo/patologia , Proteínas Hedgehog/genética , Humanos , Camundongos , Proteínas Nucleares/genética
2.
Int J Biol Macromol ; 190: 284-290, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34492245

RESUMO

Staphylococcal enterotoxin C2 (SEC2), a classical representative of superantigens, activates T cells that produce massive cytokines. This characteristic makes SEC2 a promising candidate drug for cancer immunotherapy. Previous study showed that ST-4, a SEC2 mutant, enhanced recognition of mouse T-cell receptor Vß regions, and activated the increased number of T cells that produced more cytokines. However, the underlying molecular mechanism for stimulation of human peripheral blood mononuclear cells (PBMCs) and antitumor effect on human tumor cells remains unknown. Herein, we showed that ST-4 significantly activated TCR Vß 12, 13A, 14, 15, 17, and 20 CD4+ and CD8+ T cells, which produced substantial amounts of granzyme B and perforin. These cytokines exhibited antitumor effect on K562 cells by promoting apoptosis and inducing S-phase cell cycle arrest. Conversely, the granzyme B inhibitor or perforin inhibitor significantly weakened antitumor effect of ST-4, accompanied by a decrease of cleaved proapoptotic BAX and cytochrome c, and an increase of antiapoptotic BCL2. Taken together, these data suggest that granzyme B and perforin produced by ST-4-activated CD4+ T cells and CD8+ T cells play a pivotal role in inducing K562 cell apoptosis by the mitochondrial apoptotic pathway, and support ST-4 as a potential candidate for cancer immunotherapy.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Enterotoxinas/genética , Granzimas/metabolismo , Leucemia/patologia , Mitocôndrias/metabolismo , Perforina/metabolismo , Proliferação de Células , Humanos , Células K562 , Leucemia/imunologia , Ativação Linfocitária , Mutação/genética , Receptores de Antígenos de Linfócitos T/metabolismo
3.
Front Oncol ; 11: 655630, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34136392

RESUMO

Gastric cancer is the most common malignant tumor in the digestive tract, with very high morbidity and mortality in developing countries. The pathogenesis of gastric cancer is a complex biological process mediated by abnormal regulation of proto-oncogenes and tumor suppressor genes. Although there have been some in-depth studies on gastric cancer at the molecular level, the specific mechanism has not been fully elucidated. RB family proteins (including RB, p130, and p107) are involved in cell cycle regulation, a process that largely depends on members of the E2F gene family that encode transcriptional activators and repressors. In gastric cancer, inactivation of the RB-E2F pathway serves as a core transcriptional mechanism that drives cell cycle progression, and is regulated by cyclins, cyclin-dependent kinases, cyclin-dependent kinase inhibitors, p53, Helicobacter pylori and some other upstream molecules. The E2F proteins are encoded by eight genes (i.e. E2F1 to E2F8), each of which may play a specific role in gastric cancer. Interestingly, a single E2F such as E2F1 can activate or repress transcription, and enhance or inhibit cell proliferation, depending on the cell environment. Thus, the function of the E2F transcription factor family is very complex and needs further exploration. Importantly, the presence of H. pylori in stomach mucosa may affect the RB and p53 tumor suppressor systems, thereby promoting the occurrence of gastric cancer. This review aims to summarize recent research progress on important roles of the complex RB-E2F signaling network in the development and effective treatment of gastric cancer.

4.
Oncogene ; 34(43): 5436-5446, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25639876

RESUMO

DNA methyltransferase 3A (DNMT3A) catalyzes cytosine methylation of mammalian genomic DNA. In addition to myeloid malignancies, mutations in DNMT3A have been recently reported in T-cell lymphoma and leukemia, implying a possible involvement in the pathogenesis of human diseases. However, the role of Dnmt3a in T-cell transformation in vivo is poorly understood. Here we analyzed the functional consequences of Dnmt3a inactivation in a mouse model of MYC-induced T-cell lymphomagenesis (MTCL). Loss of Dnmt3a delayed tumorigenesis by suppressing cellular proliferation during disease progression. Gene expression profiling and pathway analysis identified upregulation of 17 putative tumor suppressor genes, including DNA methyltransferase Dnmt3b, in Dnmt3a-deficient lymphomas as molecular events potentially responsible for the delayed lymphomagenesis in Dnmt3a(Δ/Δ) mice. Interestingly, promoter and gene body methylation of these genes was not substantially changed between control and Dnmt3a-deficient lymphomas, suggesting that Dnmt3a may inhibit their expression in a methylation-independent manner. Re-expression of both wild type and catalytically inactive Dnmt3a in Dnmt3a(Δ/Δ) lymphoma cells in vitro inhibited Dnmt3b expression, indicating that Dnmt3b upregulation may be directly repressed by Dnmt3a. Importantly, genetic inactivation of Dnmt3b accelerated lymphomagenesis in Dnmt3a(Δ/Δ) mice, demonstrating that upregulation of Dnmt3b is a relevant molecular change in Dnmt3a-deficient lymphomas that inhibits disease progression. Collectively, our data demonstrate an unexpected oncogenic role for Dnmt3a in MTCL through methylation-independent repression of Dnmt3b and possibly other tumor suppressor genes.


Assuntos
Carcinogênese/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Linfoma de Células T/genética , Linfoma de Células T/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Proliferação de Células/genética , DNA/genética , DNA Metiltransferase 3A , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Regiões Promotoras Genéticas/genética , Transcriptoma/genética , Regulação para Cima/genética , DNA Metiltransferase 3B
5.
PLoS One ; 9(6): e99525, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24937171

RESUMO

Despite all the blood-based biomarkers used to monitor prostate cancer patients, prostate cancer remains as the second common cause of cancer mortality in men in the United States. This is largely due to a lack of understanding of the molecular pathways that are responsible for the aggressive forms of prostate cancers, the castrate-resistant prostate cancer and the metastatic prostate cancer. Cell signaling pathways activated by the ERBB2 oncogene or the RAS oncogene are frequently found to be altered in metastatic prostate cancers. To evaluate and define the role of the ERBB2/RAS pathway in prostate cancer metastasis, we have evaluated the impact of ERBB2- or RAS-overexpression on the metastatic potentials for four prostate cancer cell lines derived from tumors with different androgen sensitivities. To do so, we transfected the human DU145, LnCaP, and PC3 prostate cancer cells and the murine Myc-CaP prostate cancer cells with the activated form of ERBB2 or H-RAS and assessed their metastatic potentials by three complementary assays, a wound healing assay, a transwell motility assay, and a transwell invasion assay. We showed that while overexpression of ERBB2 increased the metastatic potential of the androgen-insensitive prostate cancer cells (i.e. PC3 and DU145), it did not affect metastatic potentials of the androgen-sensitive prostate cancer cells (i.e. LnCaP and Myc-CaP). In contrast, overexpression of H-RAS only increased the cell motility of Myc-CaP cells, which overexpress the human c-MYC oncogene. Our data suggest that ERBB2 collaborates with androgen signaling to promote prostate cancer metastasis, and that although RAS is one of the critical downstream effectors of ERBB2, it does not phenocopy ERBB2 for its impact on the metastatic potentials of prostate cancer cell lines.


Assuntos
Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptor ErbB-2/fisiologia , Androgênios/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Senescência Celular , Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Metástase Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
6.
Mol Cell Biol ; 34(15): 2833-47, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24865965

RESUMO

Rb is critical for promoting cell cycle exit in cells undergoing terminal differentiation. Here we show that during erythroid terminal differentiation, Rb plays a previously unappreciated and unorthodox role in promoting DNA replication and cell cycle progression. Specifically, inactivation of Rb in erythroid cells led to stressed DNA replication, increased DNA damage, and impaired cell cycle progression, culminating in defective terminal differentiation and anemia. Importantly, all of these defects associated with Rb loss were exacerbated by the concomitant inactivation of E2f8. Gene expression profiling and chromatin immunoprecipitation (ChIP) revealed that Rb and E2F8 cosuppressed a large array of E2F target genes that are critical for DNA replication and cell cycle progression. Remarkably, inactivation of E2f2 rescued the erythropoietic defects resulting from Rb and E2f8 deficiencies. Interestingly, real-time quantitative PCR (qPCR) on E2F2 ChIPs indicated that inactivation of Rb and E2f8 synergizes to increase E2F2 binding to its target gene promoters. Taken together, we propose that Rb and E2F8 collaborate to promote DNA replication and erythroid terminal differentiation by preventing E2F2-mediated aberrant transcriptional activation through the ability of Rb to bind and sequester E2F2 and the ability of E2F8 to compete with E2F2 for E2f-binding sites on target gene promoters.


Assuntos
Diferenciação Celular/genética , Replicação do DNA/genética , Células Eritroides/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retinoblastoma/genética , Retinoblastoma/metabolismo , Animais , Sítios de Ligação/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Eritropoese/genética , Perfilação da Expressão Gênica/métodos , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Ativação Transcricional/genética
7.
Mol Cell Biol ; 33(21): 4321-33, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24001767

RESUMO

DNA cytosine methylation is an epigenetic modification involved in the transcriptional repression of genes controlling a variety of physiological processes, including hematopoiesis. DNA methyltransferase 1 (Dnmt1) is a key enzyme involved in the somatic inheritance of DNA methylation and thus plays a critical role in epigenomic stability. Aberrant methylation contributes to the pathogenesis of human cancer and of hematologic malignancies in particular. To gain deeper insight into the function of Dnmt1 in lymphoid malignancies, we genetically inactivated Dnmt1 in a mouse model of MYC-induced T-cell lymphomagenesis. We show that loss of Dnmt1 delays lymphomagenesis by suppressing normal hematopoiesis and impairing tumor cell proliferation. Acute inactivation of Dnmt1 in primary lymphoma cells rapidly induced apoptosis, indicating that Dnmt1 is required to sustain T-cell lymphomas. Using high-resolution genome-wide profiling, we identified differentially methylated regions between control and Dnmt1-deficient lymphomas, demonstrating a locus-specific function for Dnmt1 in both maintenance and de novo promoter methylation. Dnmt1 activity is independent of the presence of Dnmt3a or Dnmt3b in de novo promoter methylation of the H2-Ab1 gene. Collectively, these data show for the first time that Dnmt1 is critical for the prevention and maintenance of T-cell lymphomas and contributes to aberrant methylation by both de novo and maintenance methylation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Linfoma de Células T/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Carcinogênese/genética , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Hematopoese , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Linfócitos T/fisiologia , Transcrição Gênica , Transcriptoma
8.
Blood ; 119(19): 4532-42, 2012 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-22422820

RESUMO

The retinoblastoma (Rb) tumor suppressor plays important roles in regulating hematopoiesis, particularly erythropoiesis. In an effort to understand whether Rb function can be mediated by E2F transcription factors in a BM-derived hematopoietic system in mice, we uncovered a functional synergy between Rb and E2F8 to promote erythropoiesis and to prevent anemia. Specifically, whereas Mx1-Cre-mediated inactivation of Rb or E2f8 in hematopoietic stem cells only led to mild erythropoietic defects, concomitant inactivation of both genes resulted in marked ineffective erythropoiesis and mild hemolysis, leading to severe anemia despite the presence of enhanced extramedullary erythropoiesis. Interestingly, although ineffective erythropoiesis was already present in the RbΔ/Δ mice and exacerbated in the RbΔ/Δ;E2f8Δ/Δ mice, hemolysis was exclusively manifested in the double-knockout mice. Using an adoptive transfer system and an erythroid-specific knockout system, we have shown that the synergy of Rb and E2f8 deficiency in triggering severe anemia is intrinsic to the erythroid lineage. Surprisingly, concomitant inactivation of Rb and E2f7, a close family member of E2f8, did not substantially worsen the erythropoietic defect resulted from Rb deficiency. The results of the present study reveal the specificity of E2F8 in mediating Rb function in erythropoiesis and suggest critical and overlapping roles of Rb and E2f8 in maintaining normal erythropoiesis and in preventing hemolysis.


Assuntos
Anemia/genética , Inativação Gênica/fisiologia , Genes do Retinoblastoma/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas Repressoras/genética , Anemia/metabolismo , Anemia/patologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo/genética , Epistasia Genética/fisiologia , Células Eritroides/metabolismo , Eritropoese/genética , Eritropoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Hemólise/genética , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/fisiologia , Índice de Gravidade de Doença
9.
Mol Cell Biol ; 29(2): 414-24, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19015245

RESUMO

The E2f3 locus encodes two Rb-binding gene products, E2F3a and E2F3b, which are differentially regulated during the cell cycle and are thought to be critical for cell cycle progression. We targeted the individual inactivation of E2f3a or E2f3b in mice and examined their contributions to cell proliferation and development. Chromatin immunoprecipitation and gene expression experiments using mouse embryo fibroblasts deficient in each isoform showed that E2F3a and E2F3b contribute to G(1)/S-specific gene expression and cell proliferation. Expression of E2f3a or E2f3b was sufficient to support E2F target gene expression and cell proliferation in the absence of other E2F activators, E2f1 and E2f2, suggesting that these isoforms have redundant functions. Consistent with this notion, E2f3a(-/-) and E2f3b(-/-) embryos developed normally, whereas embryos lacking both isoforms (E2f3(-/-)) died in utero. We also find that E2f3a and E2f3b have redundant and nonredundant roles in the context of Rb mutation. Analysis of double-knockout embryos suggests that the ectopic proliferation and apoptosis in Rb(-/-) embryos is mainly mediated by E2f3a in the placenta and nervous system and by both E2f3a and E2f3b in lens fiber cells. Together, we conclude that the contributions of E2F3a and E2F3b in cell proliferation and development are context dependent.


Assuntos
Proliferação de Células , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/fisiologia , Desenvolvimento Embrionário , Genes do Retinoblastoma , Animais , Linhagem Celular , Sistema Nervoso Central/embriologia , Imunoprecipitação da Cromatina , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário/genética , Eritropoese/genética , Olho/embriologia , Feminino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Sistema Nervoso Periférico/embriologia , Placenta/metabolismo , Placenta/patologia , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Ativação Transcricional , Trofoblastos/citologia , Trofoblastos/fisiologia
10.
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
11.
Genes Dev ; 21(1): 85-97, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210791

RESUMO

The inactivation of the retinoblastoma (Rb) tumor suppressor gene in mice results in ectopic proliferation, apoptosis, and impaired differentiation in extraembryonic, neural, and erythroid lineages, culminating in fetal death by embryonic day 15.5 (E15.5). Here we show that the specific loss of Rb in trophoblast stem (TS) cells, but not in trophoblast derivatives, leads to an overexpansion of trophoblasts, a disruption of placental architecture, and fetal death by E15.5. Despite profound placental abnormalities, fetal tissues appeared remarkably normal, suggesting that the full manifestation of fetal phenotypes requires the loss of Rb in both extraembryonic and fetal tissues. Loss of Rb resulted in an increase of E2f3 expression, and the combined ablation of Rb and E2f3 significantly suppressed Rb mutant phenotypes. This rescue appears to be cell autonomous since the inactivation of Rb and E2f3 in TS cells restored placental development and extended the life of embryos to E17.5. Taken together, these results demonstrate that loss of Rb in TS cells is the defining event causing lethality of Rb(-/-) embryos and reveal the convergence of extraembryonic and fetal functions of Rb in neural and erythroid development. We conclude that the Rb pathway plays a critical role in the maintenance of a mammalian stem cell population.


Assuntos
Placenta/metabolismo , Proteína do Retinoblastoma/fisiologia , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Apoptose , Diferenciação Celular , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feto/citologia , Feto/embriologia , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação/genética , Fenótipo , Placenta/anormalidades , Placenta/citologia , Proteína do Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Trofoblastos/metabolismo
12.
Mol Cell Biol ; 27(1): 65-78, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17167174

RESUMO

E2F-mediated control of gene expression is believed to have an essential role in the control of cellular proliferation. Using a conditional gene-targeting approach, we show that the targeted disruption of the entire E2F activator subclass composed of E2f1, E2f2, and E2f3 in mouse embryonic fibroblasts leads to the activation of p53 and the induction of p53 target genes, including p21(CIP1). Consequently, cyclin-dependent kinase activity and retinoblastoma (Rb) phosphorylation are dramatically inhibited, leading to Rb/E2F-mediated repression of E2F target gene expression and a severe block in cellular proliferation. Inactivation of p53 in E2f1-, E2f2-, and E2f3-deficient cells, either by spontaneous mutation or by conditional gene ablation, prevented the induction of p21(CIP1) and many other p53 target genes. As a result, cyclin-dependent kinase activity, Rb phosphorylation, and E2F target gene expression were restored to nearly normal levels, rendering cells responsive to normal growth signals. These findings suggest that a critical function of the E2F1, E2F2, and E2F3 activators is in the control of a p53-dependent axis that indirectly regulates E2F-mediated transcriptional repression and cellular proliferation.


Assuntos
Fator de Transcrição E2F1/fisiologia , Fator de Transcrição E2F2/fisiologia , Fator de Transcrição E2F3/fisiologia , Regulação da Expressão Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Proliferação de Células , Fibroblastos/metabolismo , Marcação de Genes , Camundongos , Camundongos Knockout , Mutação , Fosforilação , Proteína do Retinoblastoma/metabolismo , Transcrição Gênica
13.
J Clin Invest ; 116(10): 2707-16, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16981007

RESUMO

We previously identified a rearrangement of mixed-lineage leukemia (MLL) gene (also known as ALL-1, HRX, and HTRX1), consisting of an in-frame partial tandem duplication (PTD) of exons 5 through 11 in the absence of a partner gene, occurring in approximately 4%-7% of patients with acute myeloid leukemia (AML) and normal cytogenetics, and associated with a poor prognosis. The mechanism by which the MLL PTD contributes to aberrant hematopoiesis and/or leukemia is unknown. To examine this, we generated a mouse knockin model in which exons 5 through 11 of the murine Mll gene were targeted to intron 4 of the endogenous Mll locus. Mll(PTD/WT) mice exhibit an alteration in the boundaries of normal homeobox (Hox) gene expression during embryogenesis, resulting in axial skeletal defects and increased numbers of hematopoietic progenitor cells. Mll(PTD/WT) mice overexpress Hoxa7, Hoxa9, and Hoxa10 in spleen, BM, and blood. An increase in histone H3/H4 acetylation and histone H3 lysine 4 (Lys4) methylation within the Hoxa7 and Hoxa9 promoters provides an epigenetic mechanism by which this overexpression occurs in vivo and an etiologic role for MLL PTD gain of function in the genesis of AML.


Assuntos
Epigênese Genética/genética , Duplicação Gênica , Expressão Gênica/genética , Proteínas de Homeodomínio/genética , Proteína de Leucina Linfoide-Mieloide/genética , Sequências de Repetição em Tandem/genética , Animais , Apoptose/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Rearranjo Gênico/genética , Genótipo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Proteínas Homeobox A10 , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Fenótipo , Regiões Promotoras Genéticas/genética , Costelas/anormalidades , Costelas/metabolismo
14.
Nat Genet ; 36(4): 351-60, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14991054

RESUMO

The retinoblastoma protein (Rb) regulates proliferation, cell fate specification and differentiation in the developing central nervous system (CNS), but the role of Rb in the developing mouse retina has not been studied, because Rb-deficient embryos die before the retinas are fully formed. We combined several genetic approaches to explore the role of Rb in the mouse retina. During postnatal development, Rb is expressed in proliferating retinal progenitor cells and differentiating rod photoreceptors. In the absence of Rb, progenitor cells continue to divide, and rods do not mature. To determine whether Rb functions in these processes in a cell-autonomous manner, we used a replication-incompetent retrovirus encoding Cre recombinase to inactivate the Rb1(lox) allele in individual retinal progenitor cells in vivo. Combined with data from studies of conditional inactivation of Rb1 using a combination of Cre transgenic mouse lines, these results show that Rb is required in a cell-autonomous manner for appropriate exit from the cell cycle of retinal progenitor cells and for rod development.


Assuntos
Divisão Celular/fisiologia , Retina/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Proteína do Retinoblastoma/fisiologia , Animais , Separação Celular , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Camundongos , Retina/crescimento & desenvolvimento
15.
Proc Natl Acad Sci U S A ; 100(11): 6546-51, 2003 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12732721

RESUMO

Retinoblastoma (Rb)-deficient embryos show severe defects in neurogenesis, erythropoiesis, and lens development and die at embryonic day 14.5. Our recent results demonstrated a drastic disorganization of the labyrinth layer in the placenta of Rb-deficient embryos, accompanied by reduced placental transport function. When these Rb-/- embryos were supplied with a wild-type placenta by using either tetraploid aggregation or genetic approaches, animals survived until birth. Here we analyze the role of extraembryonic Rb in regulating proliferation, apoptosis, and differentiation in the rescued animals at different developmental stages. Many of the neurological and erythroid abnormalities thought to be responsible for the embryonic lethality of Rb-/- animals, including the ectopic apoptosis in the CNS, were virtually absent in rescued Rb-/- pups. However, rescued animals died at birth with severe skeletal muscle defects. Like in Rb knockout embryos, rescued animals showed a marked increase in DNA replication and cell division in the CNS. In sharp contrast, the typical widespread neuronal apoptosis was absent in Rb-deficient embryos reconstituted with a normal placenta. In lens fiber cells, however, the inappropriate proliferation and apoptosis that is normally observed in Rb-/- embryos continued unabated in rescued animals. These results demonstrate that Rb function in extraembryonic lineages plays an important role in the survival of neuronal cells and in the differentiation of the erythroid lineage, providing mechanistic insight into the cell autonomous and nonautonomous functions of Rb during development.


Assuntos
Apoptose/fisiologia , Sistema Nervoso Central/citologia , Proteína do Retinoblastoma/fisiologia , Animais , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína do Retinoblastoma/genética
16.
Nature ; 421(6926): 942-7, 2003 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-12607001

RESUMO

The retinoblastoma (Rb) gene was the first tumour suppressor identified. Inactivation of Rb in mice results in unscheduled cell proliferation, apoptosis and widespread developmental defects, leading to embryonic death by day 14.5 (refs 2-4). However, the actual cause of the embryonic lethality has not been fully investigated. Here we show that loss of Rb leads to excessive proliferation of trophoblast cells and a severe disruption of the normal labyrinth architecture in the placenta. This is accompanied by a decrease in vascularization and a reduction in placental transport function. We used two complementary techniques-tetraploid aggregation and conditional knockout strategies-to demonstrate that Rb-deficient embryos supplied with a wild-type placenta can be carried to term, but die soon after birth. Most of the neurological and erythroid abnormalities thought to be responsible for the embryonic lethality of Rb-null animals were virtually absent in rescued Rb-null pups. These findings identify and define a key function of Rb in extra-embryonic cell lineages that is required for embryonic development and viability, and provide a mechanism for the cell autonomous versus non-cell autonomous roles of Rb in development.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Placenta/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Divisão Celular , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/citologia , Ácidos Graxos/metabolismo , Feminino , Feto/irrigação sanguínea , Feto/citologia , Feto/embriologia , Feto/metabolismo , Deleção de Genes , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Placenta/anormalidades , Placenta/irrigação sanguínea , Placenta/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína do Retinoblastoma/genética , Trofoblastos/citologia , Trofoblastos/metabolismo
17.
Cell Growth Differ ; 13(5): 215-25, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12065245

RESUMO

The Rb/E2F pathway plays a critical role in the control ofcellular proliferation. Here, we report that E2F1, E2F2, and E2F3 make major individual contributions toward the in vivo phenotypic consequences of Rb deficiency. In the developing lens of Rb(-/-) embryos, loss of E2F1, E2F2, or E2F3 reduces the unscheduled proliferation of fiber cells, with the loss of E2F3 having the most pronounced effect. In Rb-deficient retinas, all three E2Fs contribute equally to the ectopic proliferation of postmitotic neuronal cells. In contrast, E2F1 is unique in mediating apoptosis in both Rb(-/-) lenses and retinas. In the central nervous system, loss of E2F1 or E2F3 can almost completely eliminate the ectopic DNA replication and apoptosis observed in Rb(-/-) embryos, and loss of E2F2 partially reduces the unscheduled DNA replication and has no effect on apoptosis. These results provide clear evidence for functional specificity among E2Fs in the control of Rb-dependent proliferation and apoptosis in a tissue-specific manner.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Regulação da Expressão Gênica no Desenvolvimento , Retina/embriologia , Proteína do Retinoblastoma/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose/fisiologia , Divisão Celular/fisiologia , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F2 , Fator de Transcrição E2F3 , Feminino , Morte Fetal , Camundongos , Camundongos Knockout , Fenótipo , Gravidez , Retina/citologia , Retina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...