Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
Leukemia ; 28(3): 577-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24080946

RESUMO

Histone deacetylase (HDAC) inhibitors (HDACis) are well-characterized anti-cancer agents with promising results in clinical trials. However, mechanistically little is known regarding their selectivity in killing malignant cells while sparing normal cells. Gene expression-based chemical genomics identified HDACis as being particularly potent against Down syndrome-associated myeloid leukemia (DS-AMKL) blasts. Investigating the antileukemic function of HDACis revealed their transcriptional and post-translational regulation of key autophagic proteins, including ATG7. This leads to suppression of autophagy, a lysosomal degradation process that can protect cells against damaged or unnecessary organelles and protein aggregates. DS-AMKL cells exhibit low baseline autophagy due to mammalian target of rapamycin (mTOR) activation. Consequently, HDAC inhibition repressed autophagy below a critical threshold, which resulted in accumulation of mitochondria, production of reactive oxygen species, DNA damage and apoptosis. Those HDACi-mediated effects could be reverted upon autophagy activation or aggravated upon further pharmacological or genetic inhibition. Our findings were further extended to other major acute myeloid leukemia subgroups with low basal level autophagy. The constitutive suppression of autophagy due to mTOR activation represents an inherent difference between cancer and normal cells. Thus, via autophagy suppression, HDACis deprive cells of an essential pro-survival mechanism, which translates into an attractive strategy to specifically target cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Leucemia Mieloide/patologia , Animais , Humanos , Leucemia Mieloide/imunologia , Leucemia Mieloide/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Oncogene ; 31(18): 2270-82, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21927024

RESUMO

Tumors are thought to be sustained by a reservoir of self-renewing cells, termed tumor-initiating cells or cancer stem cells. Osteosarcomas are high-grade sarcomas derived from osteoblast progenitor cells and are the most common pediatric bone malignancy. In this report we show that the stem cell transcription factor Sox2 is highly expressed in human and murine osteosarcoma (mOS) cell lines as well as in the tumor samples. Osteosarcoma cells have increased ability to grow in suspension as osteospheres, that are greatly enriched in expression of Sox2 and the stem cell marker, Sca-1. Depletion of Sox2 by short-hairpin RNAs in independent mOS-derived cells drastically reduces their transformed properties in vitro and their ability to form tumors. Sox2-depleted osteosarcoma cells can no longer form osteospheres and differentiate into mature osteoblasts. Concomitantly, they exhibit decreased Sca-1 expression and upregulation of the Wnt signaling pathway. Thus, despite other mutations, these cells maintain a requirement for Sox2 for tumorigenicity. Our data indicate that Sox2 is required for osteosarcoma cell self renewal, and that Sox2 antagonizes the pro-differentiation Wnt pathway that can in turn reduce Sox2 expression. These studies define Sox2 as a survival factor and a novel biomarker of self renewal in osteosarcomas, and support a tumor suppressive role for the Wnt pathway in tumors of mesenchymal origin. Our findings could provide the basis for novel therapeutic strategies based on inhibiting Sox2 or enhancing Wnt signaling for the treatment of osteosarcomas.


Assuntos
Neoplasias Ósseas/genética , Proliferação de Células , Células-Tronco Neoplásicas , Osteossarcoma/genética , Fatores de Transcrição SOXB1/genética , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Neoplasias Ósseas/patologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Osteossarcoma/patologia , Transdução de Sinais , Via de Sinalização Wnt
3.
Nature ; 467(7313): 285-90, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20644535

RESUMO

Somatic cell nuclear transfer and transcription-factor-based reprogramming revert adult cells to an embryonic state, and yield pluripotent stem cells that can generate all tissues. Through different mechanisms and kinetics, these two reprogramming methods reset genomic methylation, an epigenetic modification of DNA that influences gene expression, leading us to hypothesize that the resulting pluripotent stem cells might have different properties. Here we observe that low-passage induced pluripotent stem cells (iPSCs) derived by factor-based reprogramming of adult murine tissues harbour residual DNA methylation signatures characteristic of their somatic tissue of origin, which favours their differentiation along lineages related to the donor cell, while restricting alternative cell fates. Such an 'epigenetic memory' of the donor tissue could be reset by differentiation and serial reprogramming, or by treatment of iPSCs with chromatin-modifying drugs. In contrast, the differentiation and methylation of nuclear-transfer-derived pluripotent stem cells were more similar to classical embryonic stem cells than were iPSCs. Our data indicate that nuclear transfer is more effective at establishing the ground state of pluripotency than factor-based reprogramming, which can leave an epigenetic memory of the tissue of origin that may influence efforts at directed differentiation for applications in disease modelling or treatment.


Assuntos
Epigênese Genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Reprogramação Celular/genética , Metilação de DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genoma/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Técnicas de Transferência Nuclear , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Artigo em Inglês | MEDLINE | ID: mdl-19478325

RESUMO

Embryonic stem (ES) cells are capable of continuous self-renewal and pluripotential differentiation. A "core" set of transcription factors, Oct4, Sox2, and Nanog, maintains the ES cell state, whereas various combinations of factors, invariably including Oct4 and Sox2, reprogram somatic cells to pluripotency. We have sought to define the transcriptional network controlling pluripotency in mouse ES cells through combined proteomic and genomic approaches. We constructed a protein interaction network surrounding Nanog and determined gene targets of the core and reprogramming factors, plus others. The expanded transcriptional network we have constructed forms the basis for further studies of directed differentiation and lineage reprogramming, and a paradigm for comprehensive elucidation of regulatory pathways in other stem cells.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Transcrição Gênica , Animais , Linhagem Celular , Redes Reguladoras de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Camundongos , Modelos Biológicos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/fisiologia , Regiões Promotoras Genéticas , Proteômica , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
5.
EMBO J ; 20(23): 6909-18, 2001 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-11726526

RESUMO

Although the physiological role of tissue-specific translational control of gene expression in mammals has long been suspected on the basis of biochemical studies, direct evidence has been lacking. Here, we report on the targeted disruption of the gene encoding the heme-regulated eIF2alpha kinase (HRI) in mice. We establish that HRI, which is expressed predominantly in erythroid cells, regulates the synthesis of both alpha- and beta-globins in red blood cell (RBC) precursors by inhibiting the general translation initiation factor eIF2. This inhibition occurs when the intracellular concentration of heme declines, thereby preventing the synthesis of globin peptides in excess of heme. In iron-deficient HRI(-/-) mice, globins devoid of heme aggregated within the RBC and its precursors, resulting in a hyperchromic, normocytic anemia with decreased RBC counts, compensatory erythroid hyperplasia and accelerated apoptosis in bone marrow and spleen. Thus, HRI is a physiological regulator of gene expression and cell survival in the erythroid lineage.


Assuntos
Eritrócitos/citologia , Eritrócitos/enzimologia , Regulação Enzimológica da Expressão Gênica , Deficiências de Ferro , Biossíntese de Proteínas , eIF-2 Quinase/metabolismo , eIF-2 Quinase/fisiologia , Animais , Apoptose , Northern Blotting , Western Blotting , Linhagem da Célula , Separação Celular , Sobrevivência Celular , Clonagem Molecular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Fator de Iniciação 2 em Eucariotos/metabolismo , Citometria de Fluxo , Biblioteca Gênica , Genótipo , Heme/biossíntese , Ferro/metabolismo , Camundongos , Microscopia Eletrônica , Modelos Biológicos , Fosforilação , Polirribossomos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Protoporfirinas/biossíntese , Reticulócitos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico , Fatores de Tempo
6.
Mol Cell Biol ; 21(24): 8547-64, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713289

RESUMO

E2F activity is critical for the control of the G(1) to S phase transition. We show that the combined loss of E2F1 and E2F2 results in profound effects on hematopoietic cell proliferation and differentiation, as well as increased tumorigenesis and decreased lymphocyte tolerance. The loss of E2F1 and E2F2 impedes B-cell differentiation, and hematopoietic progenitor cells in the bone marrow of mice lacking E2F1 and E2F2 exhibit increased cell cycling. Importantly, we show that E2F1 and E2F2 double-knockout T cells exhibit more rapid entry into S phase following antigenic stimulation. Furthermore, T cells lacking E2F1 and E2F2 proliferate much more extensively in response to subthreshold antigenic stimulation. Consistent with these observations, E2F1/E2F2 mutant mice are highly predisposed to the development of tumors, and some mice exhibit signs of autoimmunity.


Assuntos
Antígenos/metabolismo , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Neoplasias/metabolismo , Linfócitos T/citologia , Linfócitos T/fisiologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Fatores Etários , Animais , Western Blotting , Bromodesoxiuridina/metabolismo , Diferenciação Celular , Divisão Celular , Células Cultivadas , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F2 , Feminino , Citometria de Fluxo , Genótipo , Linfonodos/citologia , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Ribonucleases/metabolismo , Baço/citologia , Fatores de Tempo
7.
J Exp Med ; 194(10): 1461-71, 2001 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-11714753

RESUMO

The development of naive CD4+ T cells into a T helper (Th) 2 subset capable of producing interleukin (IL)-4, IL-5, and IL-13 involves a signal transducer and activator of transcription (Stat)6-dependent induction of GATA-3 expression, followed by Stat6-independent GATA-3 autoactivation. The friend of GATA (FOG)-1 protein regulates GATA transcription factor activity in several stages of hematopoietic development including erythrocyte and megakaryocyte differentiation, but whether FOG-1 regulates GATA-3 in T cells is uncertain. We show that FOG-1 can repress GATA-3-dependent activation of the IL-5 promoter in T cells. Also, FOG-1 overexpression during primary activation of naive T cells inhibited Th2 development in CD4+ T cells. FOG-1 fully repressed GATA-3-dependent Th2 development and GATA-3 autoactivation, but not Stat6-dependent induction of GATA-3. FOG-1 overexpression repressed development of Th2 cells from naive T cells, but did not reverse the phenotype of fully committed Th2 cells. Thus, FOG-1 may be one factor capable of regulating the Th2 development.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Proteínas de Transporte/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Transativadores/fisiologia , Animais , Fator de Transcrição GATA3 , Interleucina-4/farmacologia , Camundongos , Camundongos Transgênicos , Fator de Transcrição STAT6 , Células Th2/fisiologia , Fatores de Transcrição , Transcrição Gênica
8.
Curr Opin Genet Dev ; 11(5): 513-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11532392

RESUMO

Over the past year, significant new insights have been gained in our understanding of the lineage determination of red blood cells. In particular, evidence has emerged demonstrating that cross-antagonism of lineage-specific transcription factors plays an important role in determining cell phenotype by actively repressing alternate lineage gene programs.


Assuntos
Linhagem da Célula , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Humanos , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo
9.
Blood ; 98(7): 2248-55, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11568013

RESUMO

Patients with paroxysmal nocturnal hemoglobinuria (PNH) have blood cells deficient in glycosyl phosphatidylinositol (GPI)-linked proteins owing to a somatic mutation in the X-linked PIGA gene. To target Piga recombination to the erythroid/megakaryocytic lineage in mice, the Cre/loxP system was used, and Cre was expressed under the transcriptional regulatory sequences of GATA-1. Breeding of GATA1-cre (G) transgenic mice with mice carrying a floxed Piga (L) allele was associated with high embryonic lethality. However, double-transgenic (GL) mice that escaped early recombination looked healthy and were observed for 16 months. Flow cytometric analysis of peripheral blood cells showed that GL mice had up to 100% of red cells deficient in GPI-linked proteins. The loss of GPI-linked proteins on the cell surface occurred late in erythroid differentiation, causing a proportion of red cells to express low residual levels of GPI-linked proteins. Red cells with residual expression of GPI-linked proteins showed an intermediate sensitivity toward complement and thus resemble PNH type II cells in patients with PNH. Recombination of the floxed Piga allele was also detected in cultured megakaryocytes, mast cells, and eosinophils, but not in neutrophils, lymphocytes, or nonhematopoietic tissues. In summary, GATA1-Cre causes high-efficiency Piga gene inactivation in a GATA-1-specific pattern. For the first time, mice were generated that have almost 100% of red cells deficient in GPI-linked proteins. These animals will be valuable to further investigate the consequences of GPI-anchor deficiency on erythroid/megakaryocytic cells.


Assuntos
Proteínas de Ligação a DNA/farmacologia , Eritropoese/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Glicosilfosfatidilinositóis/deficiência , Integrases/farmacologia , Proteínas de Membrana/genética , Fatores de Transcrição/farmacologia , Proteínas Virais/farmacologia , Animais , Células da Medula Óssea/metabolismo , Linhagem da Célula , Proteínas de Ligação a DNA/fisiologia , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Hemoglobinúria Paroxística/patologia , Integrases/fisiologia , Fígado/citologia , Fígado/embriologia , Fígado/metabolismo , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Transgênicos , Recombinação Genética , Fatores de Transcrição/fisiologia , Proteínas Virais/fisiologia
10.
Development ; 128(12): 2301-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11493549

RESUMO

Hematopoietic stem cells are derived from ventral mesoderm during vertebrate development. Gene targeting experiments in the mouse have demonstrated key roles for the basic helix-loop-helix transcription factor SCL and the GATA-binding protein GATA-1 in hematopoiesis. When overexpressed in Xenopus animal cap explants, SCL and GATA-1 are each capable of specifying mesoderm to become blood. Forced expression of either factor in whole embryos, however, does not lead to ectopic blood formation. This apparent paradox between animal cap assays and whole embryo phenotype has led to the hypothesis that additional factors are involved in specifying hematopoietic mesoderm. SCL and GATA-1 interact in a transcriptional complex with the LIM domain protein LMO-2. We have cloned the Xenopus homolog of LMO-2 and show that it is expressed in a similar pattern to SCL during development. LMO-2 can specify hematopoietic mesoderm in animal cap assays. SCL and LMO-2 act synergistically to expand the blood island when overexpressed in whole embryos. Furthermore, co-expression of GATA-1 with SCL and LMO-2 leads to embryos that are ventralized and have blood throughout the dorsal-ventral axis. The synergistic effect of SCL, LMO-2 and GATA-1, taken together with the findings that these factors can form a complex in vitro, suggests that this complex specifies mesoderm to become blood during embryogenesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Eritropoese/fisiologia , Sequências Hélice-Alça-Hélice , Metaloproteínas/fisiologia , Proteínas Proto-Oncogênicas , Fatores de Transcrição/fisiologia , Proteínas de Xenopus , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Expressão Gênica , Globinas/genética , Humanos , Proteínas com Domínio LIM , Metaloproteínas/genética , Camundongos , Dados de Sequência Molecular , Proteínas Smad , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transativadores/genética , Fatores de Transcrição/genética , Xenopus laevis/embriologia
11.
Proc Natl Acad Sci U S A ; 98(13): 7342-7, 2001 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-11404479

RESUMO

Friend of GATA (FOG) proteins regulate GATA factor-activated gene transcription. During vertebrate hematopoiesis, FOG and GATA proteins cooperate to promote erythrocyte and megakaryocyte differentiation. The Drosophila FOG homologue U-shaped (Ush) is expressed similarly in the blood cell anlage during embryogenesis. During hematopoiesis, the acute myeloid leukemia 1 homologue Lozenge and Glial cells missing are required for the production of crystal cells and plasmatocytes, respectively. However, additional factors have been predicted to control crystal cell proliferation. In this report, we show that Ush is expressed in hemocyte precursors and plasmatocytes throughout embryogenesis and larval development, and the GATA factor Serpent is essential for Ush embryonic expression. Furthermore, loss of ush function results in an overproduction of crystal cells, whereas forced expression of Ush reduces this cell population. Murine FOG-1 and FOG-2 also can repress crystal cell production, but a mutant version of FOG-2 lacking a conserved motif that binds the corepressor C-terminal binding protein fails to affect the cell lineage. The GATA factor Pannier (Pnr) is required for eye and heart development in Drosophila. When Ush, FOG-1, FOG-2, or mutant FOG-2 is coexpressed with Pnr during these developmental processes, severe eye and heart phenotypes result, consistent with a conserved negative regulation of Pnr function. These results indicate that the fly and mouse FOG proteins function similarly in three distinct cellular contexts in Drosophila, but may use different mechanisms to regulate genetic events in blood vs. cardial or eye cell lineages.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila , Drosophila melanogaster/embriologia , Drosophila melanogaster/crescimento & desenvolvimento , Hemócitos/citologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/fisiologia , Olho/embriologia , Olho/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Coração/crescimento & desenvolvimento , Hemócitos/fisiologia , Larva , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Dedos de Zinco
12.
Genes Dev ; 15(7): 839-44, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11297508

RESUMO

GATA-family transcription factors are critical to the development of diverse tissues. In particular, GATA-4 has been implicated in formation of the vertebrate heart. As the mouse Gata-4 knock-out is early embryonic lethal because of a defect in ventral morphogenesis, the in vivo function of this factor in heart development remains unresolved. To search for a requirement for Gata4 in heart development, we created mice harboring a single amino acid replacement in GATA-4 that impairs its physical interaction with its presumptive cardiac cofactor FOG-2. Gata4(ki/ki) mice die just after embryonic day (E) 12.5 exhibiting features in common with Fog2(-/-) embryos as well as additional semilunar cardiac valve defects and a double-outlet right ventricle. These findings establish an intrinsic requirement for GATA-4 in heart development. We also infer that GATA-4 function is dependent on interaction with FOG-2 and, very likely, an additional FOG protein for distinct aspects of heart formation.


Assuntos
Anomalias dos Vasos Coronários/genética , Vasos Coronários/embriologia , Proteínas de Ligação a DNA/fisiologia , Coração Fetal/crescimento & desenvolvimento , Cardiopatias Congênitas/genética , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Anomalias dos Vasos Coronários/embriologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Fatores de Ligação de DNA Eritroide Específicos , Coração Fetal/patologia , Fator de Transcrição GATA4 , Genes Letais , Idade Gestacional , Cardiopatias Congênitas/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Morfogênese/genética , Mutagênese Sítio-Dirigida , Conformação Proteica , Fatores de Transcrição/biossíntese , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica , Valina/química , Proteínas de Peixe-Zebra
13.
Blood ; 97(1): 324-6, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11133778

RESUMO

Reporter mouse strains are important tools for monitoring Cre recombinase-mediated excision in vivo. In practice, excision may be incomplete in a given population due to threshold level or variegated expression of Cre. Hence, it is desirable in many experimental contexts to isolate cells that have undergone excision to assess the consequences of gene ablation. To generate alternative reporter mice, an enhanced green fluorescent protein (EGFP) gene was targeted to the retroviral-trapped ROSA26 locus. Upon Cre-mediated excision of "Stop" sequences, EGFP was expressed ubiquitously during embryogenesis and in adult tissues (including T cells, B cells, and myeloid cells). Using this new reporter strain, separation of excised from nonexcised cells in vitro was achieved in thymocytes in a noninvasive manner based on activated EGFP expression. This new EGFP reporter strain should facilitate a variety of conditional gene-targeting experiments, including the functional studies of hematopoietic cells in lineage-specific knockout mice.


Assuntos
Genes Reporter , Integrases/metabolismo , Proteínas Luminescentes/biossíntese , Proteínas/genética , Proteínas Virais , Animais , Células Sanguíneas , Embrião de Mamíferos , Regulação da Expressão Gênica , Marcação de Genes/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde , Indicadores e Reagentes , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , RNA não Traduzido , Timo/citologia
14.
Immunity ; 15(6): 959-70, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11754817

RESUMO

E2Fs are important regulators of proliferation, differentiation, and apoptosis. Here we characterize the phenotype of mice deficient in E2F2. We show that E2F2 is required for immunologic self-tolerance. E2F2(-/-) mice develop late-onset autoimmune features, characterized by widespread inflammatory infiltrates, glomerular immunocomplex deposition, and anti-nuclear antibodies. E2F2-deficient T lymphocytes exhibit enhanced TCR-stimulated proliferation and a lower activation threshold, leading to the accumulation of a population of autoreactive effector/memory T lymphocytes, which appear to be responsible for causing autoimmunity in E2F2-deficient mice. Finally, we provide support for a model to explain E2F2's unexpected role as a suppressor of T lymphocyte proliferation. Rather than functioning as a transcriptional activator, E2F2 appears to function as a transcriptional repressor of genes required for normal S phase entry, particularly E2F1.


Assuntos
Doenças Autoimunes/genética , Autoimunidade/imunologia , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/imunologia , Proteínas Repressoras/fisiologia , Tolerância a Antígenos Próprios/imunologia , Linfócitos T/citologia , Fatores de Transcrição/fisiologia , Animais , Apoptose , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Autoimunidade/genética , Divisão Celular , Quimera , Deleção Clonal , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F2 , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/imunologia , Antígeno H-Y/genética , Antígeno H-Y/imunologia , Humanos , Memória Imunológica , Inflamação , Células Jurkat , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Repressoras/genética , Fase S/genética , Tolerância a Antígenos Próprios/genética , Esplenomegalia/genética , Esplenomegalia/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Timo/imunologia , Timo/patologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção
15.
Proc Natl Acad Sci U S A ; 97(25): 13796-800, 2000 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-11095756

RESUMO

Malignant rhabdoid tumor (MRT) is an aggressive, highly lethal cancer of young children. Tumors occur in various locations, including kidney, brain, and soft tissues. Despite intensive therapy, 80% of affected children die, often within 1 year of diagnosis. The majority of MRT samples and cell lines have sustained biallelic inactivating mutations of the hSNF5 (integrase interactor 1) gene, suggesting that hSNF5 may act as a tumor suppressor. We sought to examine the role of Snf5 in development and cancer in a murine model. Here we report that Snf5 is widely expressed during embryogenesis with focal areas of high-level expression in the mandibular portion of the first branchial arch and central nervous system. Homozygous knockout of Snf5 results in embryonic lethality by embryonic day 7, whereas heterozygous mice are born at the expected frequency and appear normal. However, beginning as early as 5 weeks of age, heterozygous mice develop tumors consistent with MRT. The majority of tumors arise in soft tissues derived from the first branchial arch. Our findings constitute persuasive genetic evidence that Snf5, a core member of the Swi/Snf chromatin-remodeling complex, functions as a tumor suppressor gene, and, moreover, Snf5 heterozygotes provide a murine model of this lethal pediatric cancer.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Predisposição Genética para Doença , Tumor Rabdoide/genética , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Cromossômicas não Histona , Clonagem Molecular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteína SMARCB1 , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Fatores de Transcrição/genética
17.
Proc Natl Acad Sci U S A ; 97(25): 13696-701, 2000 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-11087841

RESUMO

Megakaryocytes lacking transcription factor GATA-1 fail to complete maturation in vivo and hyperproliferate. To define how GATA-1 regulates megakaryocyte cell growth we searched for mRNA transcripts expressed in primary wild-type, but not GATA-1(-), megakaryocytes. One differentially expressed transcript encodes inositol polyphosphate 4-phosphatase type I (4-Ptase I). This enzyme hydrolyses phosphatidylinositol 3,4-bisphosphate and also has lesser activity against soluble analogues of this lipid, inositol 3, 4-bisphosphate and inositol 1,3,4-triphosphate. Reintroduction of 4-Ptase I into both primary GATA-1(-) and wild-type megakaryocytes significantly retards cell growth, suggesting that absence of 4-Ptase I may contribute to the hyperproliferative phenotype of GATA-1(-) megakaryocytes. Overexpression of 4-Ptase I also markedly reduces growth of NIH 3T3 fibroblasts. Taken together, these data indicate that 4-Ptase I is a regulator of cell proliferation.


Assuntos
Divisão Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Fatores de Transcrição/fisiologia , Células 3T3 , Animais , Sequência de Bases , Primers do DNA , Fatores de Ligação de DNA Eritroide Específicos , Fibroblastos/citologia , Fator de Transcrição GATA1 , Megacariócitos/citologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular
18.
Genes Dev ; 14(19): 2515-25, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11018018

RESUMO

The commitment of multipotent cells to particular developmental pathways requires specific changes in their transcription factor complement to generate the patterns of gene expression characteristic of specialized cell types. We have studied the role of the GATA cofactor Friend of GATA (FOG) in the differentiation of avian multipotent hematopoietic progenitors. We found that multipotent cells express high levels of FOG mRNA, which were rapidly down-regulated upon their C/EBPbeta-mediated commitment to the eosinophil lineage. Expression of FOG in eosinophils led to a loss of eosinophil markers and the acquisition of a multipotent phenotype, and constitutive expression of FOG in multipotent progenitors blocked activation of eosinophil-specific gene expression by C/EBPbeta. Our results show that FOG is a repressor of the eosinophil lineage, and that C/EBP-mediated down-regulation of FOG is a critical step in eosinophil lineage commitment. Furthermore, our results indicate that maintenance of a multipotent state in hematopoiesis is achieved through cooperation between FOG and GATA-1. We present a model in which C/EBPbeta induces eosinophil differentiation by the coordinate direct activation of eosinophil-specific promoters and the removal of FOG, a promoter of multipotency as well as a repressor of eosinophil gene expression.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Transporte/metabolismo , Eosinófilos/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas Nucleares/metabolismo , Animais , Proteínas Aviárias , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Proteínas de Ligação a DNA , Regulação para Baixo , Eosinófilos/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Glicoproteínas de Membrana/genética , Modelos Genéticos , Células Mieloides , Fenótipo , Regiões Promotoras Genéticas , Fatores de Transcrição
19.
Blood ; 96(9): 3078-85, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11049987

RESUMO

The receptor tyrosine kinase Flk-1 is essential for embryonic blood vessel development and for tumor angiogenesis. To identify upstream transcriptional regulators of Flk-1, the gene regulatory elements that mediate endothelium-specific expression in mouse embryos were characterized. By mutational analysis, binding sites for SCL/Tal-1, GATA, and Ets transcription factors located in the Flk-1 enhancer were identified as critical elements for the endothelium-specific Flk-1 gene expression in transgenic mice. c-Ets1, a transcription factor that is coexpressed with Flk-1 during embryonic development and tumor angiogenesis, activated the Flk-1 promoter via 2 binding sites. One of these sites was required for Flk-1 promoter function in the embryonic vasculature. These results provide the first evidence that SCL/Tal-1, GATA, and Ets transcription factors act upstream of Flk-1 in a combinatorial fashion to determine embryonic blood vessel formation and are key regulators not only of the hematopoietic program, but also of vascular development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/fisiologia , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Sítios de Ligação , Bovinos , Células Cultivadas , Galinhas , Endotélio Vascular/citologia , Fatores de Ligação de DNA Eritroide Específicos , Camundongos , Dados de Sequência Molecular , Mutagênese , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas c-ets , Receptores Mitogênicos/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Mapeamento por Restrição , Deleção de Sequência , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transfecção
20.
Blood ; 96(8): 2641-8, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11023493

RESUMO

The lineage-specific transcription factors GATA-1 and PU.1 can physically interact to inhibit each other's function, but the mechanism of repression of GATA-1 function by PU.1 has not been elucidated. Both the N terminus and the C terminus of PU.1 can physically interact with the C-terminal zinc finger of GATA-1. It is demonstrated that the PU.1 N terminus, but not the C terminus, is required for inhibiting GATA-1 function. Induced overexpression of PU.1 in K562 erythroleukemia cells blocks hemin-induced erythroid differentiation. In this system, PU.1 does not affect the expression of GATA-1 messenger RNA, protein, or nuclear localization. However, GATA-1 DNA binding decreases dramatically. By means of electrophoretic mobility shift assays with purified proteins, it is demonstrated that the N-terminal 70 amino acids of PU.1 can specifically block GATA-1 DNA binding. In addition, PU.1 had a similar effect in the G1ER cell line, in which the GATA-1 null erythroid cell line G1E has been transduced with a GATA-1-estrogen receptor fusion gene, which is directly dependent on induction of the GATA-1 fusion protein to effect erythroid maturation. Consistent with in vitro binding assays, overexpression of PU.1 blocked DNA binding of the GATA-1 fusion protein as well as GATA-1-mediated erythroid differentiation of these G1ER cells. These results demonstrate a novel mechanism by which function of a lineage-specific transcription factor is inhibited by another lineage-restricted factor through direct protein-protein interactions. These findings contribute to understanding how protein-protein interactions participate in hematopoietic differentiation and leukemogenesis. (Blood. 2000;96:2641-2648)


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , DNA/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Fatores de Transcrição/antagonistas & inibidores , Transporte Biológico , Diferenciação Celular/fisiologia , Linhagem da Célula , Núcleo Celular/metabolismo , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Estradiol/farmacologia , Fator de Transcrição GATA1 , Regulação Leucêmica da Expressão Gênica/fisiologia , Genes Sintéticos , Humanos , Células K562/metabolismo , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/análise , RNA Neoplásico/análise , Receptores de Estrogênio/genética , Proteínas Recombinantes de Fusão/fisiologia , Transativadores/química , Transativadores/genética , Fatores de Transcrição/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...