Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 372(6538)2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33833093

RESUMO

DNA methylation is essential to mammalian development, and dysregulation can cause serious pathological conditions. Key enzymes responsible for deposition and removal of DNA methylation are known, but how they cooperate to regulate the methylation landscape remains a central question. Using a knockin DNA methylation reporter, we performed a genome-wide CRISPR-Cas9 screen in human embryonic stem cells to discover DNA methylation regulators. The top screen hit was an uncharacterized gene, QSER1, which proved to be a key guardian of bivalent promoters and poised enhancers of developmental genes, especially those residing in DNA methylation valleys (or canyons). We further demonstrate genetic and biochemical interactions of QSER1 and TET1, supporting their cooperation to safeguard transcriptional and developmental programs from DNMT3-mediated de novo methylation.


Assuntos
Metilação de DNA , DNA/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Sistemas CRISPR-Cas , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Genoma Humano , Humanos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , DNA Metiltransferase 3B
2.
Biol Open ; 9(6)2020 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-32580940

RESUMO

The Gata4/5/6 sub-family of zinc finger transcription factors regulate many aspects of cardiogenesis. However, critical roles in extra-embryonic endoderm also challenge comprehensive analysis during early mouse cardiogenesis, while zebrafish models have previously relied on knockdown assays. We generated targeted deletions to disrupt each gata4/5/6 gene in zebrafish and analyzed cardiac phenotypes in single, double and triple mutants. The analysis confirmed that loss of gata5 causes cardia bifida and validated functional redundancies for gata5/6 in cardiac precursor specification. Surprisingly, we discovered that gata4 is dispensable for early zebrafish development, while loss of one gata4 allele can suppress the bifid phenotype of the gata5 mutant. The gata4 mutants eventually develop an age-dependent cardiomyopathy. By combining combinations of mutant alleles, we show that cardiac specification depends primarily on an overall dosage of gata4/5/6 alleles rather than a specific gene. We also identify a specific role for gata6 in controlling ventricle morphogenesis through regulation of both the first and second heart field, while loss of both gata4/6 eliminates the ventricle. Thus, different developmental programs are dependent on total dosage, certain pairs, or specific gata4/5/6 genes during embryonic cardiogenesis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Fator de Transcrição GATA4/genética , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Organogênese/genética , Peixe-Zebra/embriologia , Alelos , Animais , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA5/metabolismo , Fator de Transcrição GATA6/metabolismo , Dosagem de Genes , Marcação de Genes , Genótipo , Morfogênese/genética , Mutação , Fenótipo
3.
Cancer Discov ; 9(5): 662-679, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30777872

RESUMO

Several lines of evidence link the canonical oncogene BCL6 to stress response. Here we demonstrate that BCL6 evolved in vertebrates as a component of the HSF1-driven stress response, which has been co-opted by the immune system to support germinal center formation and may have been decisive in the convergent evolution of humoral immunity in jawless and jawed vertebrates. We find that the highly conserved BTB corepressor binding site of BCL6 mediates stress adaptation across vertebrates. We demonstrate that pan-cancer cells hijack this stress tolerance mechanism to aberrantly express BCL6. Targeting the BCL6 BTB domain in cancer cells induces apoptosis and increases susceptibility to repeated doses of cytotoxic therapy. The chemosensitization effect upon BCL6 BTB inhibition is dependent on the derepression of TOX, implicating modulation of DNA repair as a downstream mechanism. Collectively, these data suggest a form of adaptive nononcogene addiction rooted in the natural selection of BCL6 during vertebrate evolution. SIGNIFICANCE: We demonstrate that HSF1 drives BCL6 expression to enable stress tolerance in vertebrates. We identify an HSF1-BCL6-TOX stress axis that is required by cancer cells to tolerate exposure to cytotoxic agents and points toward BCL6-targeted therapy as a way to more effectively kill a wide variety of solid tumors.This article is highlighted in the In This Issue feature, p. 565.


Assuntos
Adaptação Fisiológica/fisiologia , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Estresse Fisiológico/fisiologia , Animais , Apoptose/fisiologia , Linfócitos B/citologia , Linfócitos B/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Centro Germinativo/citologia , Centro Germinativo/fisiologia , Fatores de Transcrição de Choque Térmico/biossíntese , Fatores de Transcrição de Choque Térmico/genética , Fatores de Transcrição de Choque Térmico/metabolismo , Resposta ao Choque Térmico , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Neoplasias/enzimologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-bcl-6/genética
4.
Proc Natl Acad Sci U S A ; 112(46): E6359-68, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26578796

RESUMO

Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


Assuntos
Eritroblastos/metabolismo , Proteínas Ativadoras de GTPase/biossíntese , Regulação da Expressão Gênica , Síndromes Mielodisplásicas/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Eritroblastos/patologia , Feminino , Proteínas Ativadoras de GTPase/genética , Humanos , Masculino , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
5.
Blood ; 123(9): 1384-92, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24425803

RESUMO

The RNA-binding protein Elavl1 (also known as HuR) regulates gene expression at the posttranscriptional level. Early embryonic lethality of the mouse knockout challenges investigation into hematopoietic functions for Elavl1. We identified 2 zebrafish elavl1 genes, designated elavl1a (the predominant isoform during embryogenesis) and elavl1b. Knockdown of Elavl1a using specific morpholinos resulted in a striking loss of primitive embryonic erythropoiesis. Transcript levels for early hematopoietic regulatory genes including lmo2 and scl are unaltered, but levels of gata1 transcripts, encoding a key erythroid transcription factor, are significantly reduced in elavl1a morphants. Other mesoderm markers are mostly unchanged by depletion of Elav1a. The 3'-untranslated region (UTR) of gata1 contains putative Elavl1a-binding sites that support robust expression levels when fused to a transfected luciferase reporter gene, and Elavl1a binds the gata1 3'-UTR sequences in a manner dependent on these sites. Moreover, expression of a transgenic reporter specifically in developing embryonic erythroid cells is enhanced by addition of the gata1 3'UTR with intact Elavl1-binding sites. Injection of gata1 messenger RNA partially rescues the erythropoiesis defect caused by Elavl1 knockdown. Our study reveals a posttranscriptional regulatory mechanism by which RNA-binding protein Elavl1a regulates embryonic erythropoiesis by maintaining appropriate levels of gata1 expression.


Assuntos
Proteínas ELAV/fisiologia , Eritropoese/genética , Fator de Transcrição GATA1/genética , Processamento Pós-Transcricional do RNA/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Regiões 3' não Traduzidas/genética , Animais , Animais Geneticamente Modificados , Sítios de Ligação/genética , Linhagem Celular Tumoral , Embrião não Mamífero , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Proteínas de Peixe-Zebra/metabolismo
6.
Bioorg Med Chem ; 22(2): 673-83, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24393720

RESUMO

Retinoids comprise a group of compounds each composed of three basic parts: a trimethylated cyclohexene ring that is a bulky hydrophobic group, a conjugated tetraene side chain that functions as a linker unit, and a polar carbon-oxygen functional group. Biochemical conversion of carotenoid or other retinoids to retinoic acid (RA) is essential for normal regulation of a wide range of biological processes including development, differentiation, proliferation, and apoptosis. Retinoids regulate various physiological outputs by binding to nuclear receptors called retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which themselves are DNA-binding transcriptional regulators. The functional response of RA and their receptors are modulated by a host of coactivators and corepressors. Retinoids are essential in the development and function of several organ systems; however, deregulated retinoid signaling can contribute to serious diseases. Several natural and synthetic retinoids are in clinical use or undergoing trials for treating specific diseases including cancer. In this review, we provide a broad overview on the importance of retinoids in development and various diseases, highlighting various retinoids in the drug discovery process, ranging all the way from retinoid chemistry to clinical uses and imaging.


Assuntos
Olho/metabolismo , Rim/metabolismo , Doenças Metabólicas/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Descoberta de Drogas , Olho/química , Olho/efeitos dos fármacos , Humanos , Rim/química , Rim/efeitos dos fármacos , Doenças Metabólicas/tratamento farmacológico , Estrutura Molecular , Neoplasias/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Tretinoína/química , Tretinoína/farmacologia
7.
PLoS One ; 7(10): e46844, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056483

RESUMO

Using the zebrafish model we describe a previously unrecognized requirement for the transcription factor gata4 controlling embryonic angiogenesis. The development of a vascular plexus in the embryonic tail, the caudal hematopoietic tissue (CHT), fails in embryos depleted of gata4. Rather than forming a normal vascular plexus, the CHT of gata4 morphants remains fused, and cells in the CHT express high levels of osteogenic markers ssp1 and runx1. Definitive progenitors emerge from the hemogenic aortic endothelium, but fail to colonize the poorly vascularized CHT. We also found abnormal patterns and levels for the chemokine sdf1a in gata4 morphants, which was found to be functionally relevant, since the embryos also show defects in development of the lateral line, a mechano-sensory organ system highly dependent on a gradient of sdf1a levels. Reduction of sdf1a levels was sufficient to rescue lateral line development, circulation, and CHT morphology. The result was surprising since neither gata4 nor sdf1a is obviously expressed in the CHT. Therefore, we generated transgenic fish that conditionally express a dominant-negative gata4 isoform, and determined that gata4 function is required during gastrulation, when it is co-expressed with sdf1a in lateral mesoderm. Our study shows that the gata4 gene regulates sdf1a levels during early embryogenesis, which impacts embryonic patterning and subsequently the development of the caudal vascular plexus.


Assuntos
Vasos Sanguíneos/metabolismo , Quimiocina CXCL12/metabolismo , Fator de Transcrição GATA4/metabolismo , Peixe-Zebra/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Quimiocina CXCL12/deficiência , Quimiocina CXCL12/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fator de Transcrição GATA4/deficiência , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hematopoese , Neovascularização Fisiológica , Transdução de Sinais , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Dev Cell ; 22(3): 625-38, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22421047

RESUMO

In mammalian embryonic stem cells, the acquisition of pluripotency is dependent on Nanog, but the in vivo analysis of Nanog has been hampered by its requirement for early mouse development. In an effort to examine the role of Nanog in vivo, we identified a zebrafish Nanog ortholog and found that its knockdown impaired endoderm formation. Genome-wide transcription analysis revealed that nanog-like morphants fail to develop the extraembryonic yolk syncytial layer (YSL), which produces Nodal, required for endoderm induction. We examined the genes that were regulated by Nanog-like and identified the homeobox gene mxtx2, which is both necessary and sufficient for YSL induction. Chromatin immunoprecipitation assays and genetic studies indicated that Nanog-like directly activates mxtx2, which, in turn, specifies the YSL lineage by directly activating YSL genes. Our study identifies a Nanog-like-Mxtx2-Nodal pathway and establishes a role for Nanog-like in regulating the formation of the extraembryonic tissue required for endoderm induction.


Assuntos
Endoderma/metabolismo , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes da Sinalização Nodal/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Dados de Sequência Molecular , Proteína Homeobox Nanog , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
9.
Chem Biol Drug Des ; 73(3): 339-45, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19207470

RESUMO

We synthesized 2-substituted 2H-chromene derivatives from salicylaldehyde using potassium vinylic borates in the presence of secondary amines. Our goal was to generate novel compounds that might modulate transforming growth factor-beta signaling, based on limited rational design. Potassium vinyl trifluoroborates react with salicylaldehydes at 80 degrees C in the presence of a secondary amine and produce 2-substituted 2H-chromene derivatives with a 70-90% yield. A small library of these compounds, predicted to potentially interact with transforming growth factor-beta receptors, was screened for bioactivity in living zebrafish embryos. We found that the related compounds differentially affect development, and demonstrate one compound that produces severe body axis alterations in early embryogenesis and at lower doses affects specifically cardiovascular development. This compound modulates specifically a Smad-independent transforming growth factor-beta-regulated mitogen-activated protein kinase pathway, namely p-SAPK/JNK. These compounds, as suggested by our biological assays, may prove useful to manipulate developmental programs and develop therapeutic tools.


Assuntos
Benzopiranos/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/agonistas , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Peixe-Zebra/embriologia , Animais , Benzopiranos/síntese química , Benzopiranos/química , Avaliação Pré-Clínica de Medicamentos/métodos , Desenvolvimento Embrionário/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Bibliotecas de Moléculas Pequenas
10.
Proc Natl Acad Sci U S A ; 105(1): 94-9, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18160536

RESUMO

The ability to store fat in the form of cytoplasmic triglyceride droplets is conserved from Saccharomyces cerevisiae to humans. Although much is known regarding the composition and catabolism of lipid droplets, the molecular components necessary for the biogenesis of lipid droplets have remained obscure. Here we report the characterization of a conserved gene family important for lipid droplet formation named fat-inducing transcript (FIT). FIT1 and FIT2 are endoplasmic reticulum resident membrane proteins that induce lipid droplet accumulation in cell culture and when expressed in mouse liver. shRNA silencing of FIT2 in 3T3-LI adipocytes prevents accumulation of lipid droplets, and depletion of FIT2 in zebrafish blocks diet-induced accumulation of lipid droplets in the intestine and liver, highlighting an important role for FIT2 in lipid droplet formation in vivo. Together these studies identify and characterize a conserved gene family that is important in the fundamental process of storing fat.


Assuntos
Tecido Adiposo/fisiologia , Evolução Molecular , Regulação da Expressão Gênica , Fígado/metabolismo , Proteínas de Membrana/fisiologia , Células 3T3-L1/metabolismo , Adipócitos/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Diabetes Mellitus Experimental/metabolismo , Humanos , Lipídeos/química , Proteínas de Membrana/química , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Triglicerídeos/química , Triglicerídeos/metabolismo , Peixe-Zebra
11.
Blood ; 109(2): 516-23, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16990609

RESUMO

Bone morphogenetic protein (BMP) signaling is an important regulator of hematovascular development. However, the progenitor population that responds to BMP signaling is undefined, and the relative role of downstream mediators including Smad1 is unclear. We find that Smad1 shows a distinctive expression profile as embryonic stem (ES) cells undergo differentiation in the embryoid body (EB) system, with peak levels in cell populations enriched for the hemangioblast. To test the functional relevance of this observation, we generated an ES cell line that allows temporal control of ectopic Smad1 expression. Continuous expression of Smad1 from day 2 of EB culture does not disturb hematopoiesis, according to colony assays. In contrast, a pulse of Smad1 expression exclusively between day 2 and day 2.25 expands the population of progenitors for primitive erythroblasts and other hematopoietic lineages. This effect correlates with increased levels of transcripts encoding markers for the hemangioblast, including Runx1, Scl, and Gata2. Indeed, the pulse of Smad1 induction also expands the blast colony-forming cell (BL-CFC) population at a level that is fully sufficient to explain subsequent increases in hematopoiesis. Our data demonstrate that Smad1 expression is sufficient to expand the number of cells that commit to hemangioblast fate.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteína Smad1/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/fisiologia , Proteína Smad1/biossíntese , Proteína Smad1/genética , Fatores de Tempo
12.
Blood ; 108(9): 2989-97, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16835375

RESUMO

STAT5 regulates definitive (adult stage) erythropoiesis through its ability to transduce signals from the erythropoietin receptor. A function for STAT-dependent signaling during primitive (embryonic) erythropoiesis has not been analyzed. We tested this in the Xenopus system, because STAT5 is expressed at the right time and place to regulate development of the embryonic primitive ventral blood island. Depletion of STAT5 activity results in delayed accumulation of the first globin-expressing cells, indicating that the gene does regulate primitive erythropoiesis. Our results suggest that in this context STAT5 functions as a repressor, since forced expression of an activator isoform blocks erythropoiesis, while embryos expressing a repressor isoform develop normally. The erythroid phenotype caused by the activator isoform of STAT5 resembles that caused by overexpression of fibroblast growth factor (FGF). We show that STAT5 isoforms can function epistatic to FGF and can be phosphorylated in response to hyperactivated FGF signaling in Xenopus embryos. Therefore, our data indicate that STAT5 functions in both primitive and definitive erythropoiesis, but by different mechanisms.


Assuntos
Eritropoese/fisiologia , Fator de Transcrição STAT5/genética , Proteínas de Xenopus/genética , Xenopus/embriologia , Xenopus/genética , Animais , Embrião não Mamífero , Desenvolvimento Embrionário/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/metabolismo
13.
Biochem J ; 393(Pt 1): 311-20, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16156721

RESUMO

In a search for binding partners to Smad8, we identified the chicken homologue of the mammalian Tid1 protein (cTid1), which is a regulator of apoptosis related to the Drosophila tumour suppressor Tid56. The cTid1 coding sequence is highly conserved with mammalian Tid1, including the DnaJ domain that interacts with Hsp70 (heat-shock protein 70). The cTid1 gene is widely expressed with transcripts enriched in the developing blood islands of the embryonic-yolk sac. We show that cTid1 can bind to other members of the Smad family and that highest binding activity occurs with the negative regulatory Smad7, through the conserved MH2 domain. This interaction can have functional relevance in vivo, since co-expression of Tid1 blocks the dorsalizing and BMP (bone morphogenetic protein)-dependent regulatory activity of Smad7 in developing Xenopus embryos. The finding that these proteins can interact suggests the potential for linking two important cell survival/apoptosis pathways.


Assuntos
Proteínas Aviárias/metabolismo , Proteína Smad7/metabolismo , Proteínas de Xenopus/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Aviárias/química , Proteínas Aviárias/genética , Sítios de Ligação , Blastômeros/metabolismo , Linhagem Celular , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Xenopus laevis/embriologia , Xenopus laevis/genética , Xenopus laevis/metabolismo
14.
Nucleic Acids Res ; 33(13): 4357-67, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16061939

RESUMO

The gata2 gene encodes a transcription factor implicated in regulating early patterning of ectoderm and mesoderm, and later in numerous cell-specific gene expression programs. Activation of the gata2 gene during embryogenesis is dependent on the bone morphogenetic protein (BMP) signaling pathway, but the mechanism for how signaling controls gene activity has not been defined. We developed an assay in Xenopus embryos to analyze regulatory sequences of the zebrafish gata2 promoter that are necessary to mediate the response to BMP signaling during embryogenesis. We show that activation is Smad dependent, since it is blocked by expression of the inhibitory Smad6. Deletion analysis identified an octamer binding site that is necessary for BMP-mediated induction, and that interacts with the POU homeodomain protein Oct-1. However, this element is not sufficient to transfer a BMP response to a heterologous promoter, requiring an additional more proximal cooperating element. Based on recent studies with other BMP-dependent promoters (Drosophila vestigial and Xenopus Xvent-2), our studies of the gata2 gene suggest that POU-domain proteins comprise a common component of the BMP signaling pathway, cooperating with Smad proteins and other transcriptional activators.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regiões Promotoras Genéticas , Elementos de Resposta , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Proteína Morfogenética Óssea 4 , Fator de Transcrição GATA2 , Fator 1 de Transcrição de Octâmero , Deleção de Sequência , Transdução de Sinais , Proteínas Smad , Transativadores/metabolismo , Proteínas de Xenopus , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
J Biol Chem ; 278(7): 4705-12, 2003 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-12468531

RESUMO

Hepatocyte growth factor (HGF) is released in response to myocardial infarction and may play a role in regulating cardiac remodeling. Recently, HGF was found to inhibit the apoptosis of cardiac muscle cells. Because GATA-4 can induce cell survival, the effects of HGF on GATA-4 activity were investigated. Treatment of HL-1 cells or primary adult rat cardiac myocytes with HGF, at concentrations that can be detected in the human serum after myocardial infarction, rapidly enhances GATA-4 DNA-binding activity. The enhanced DNA-binding activity is associated with the phosphorylation of GATA-4. HGF-induced phosphorylation and activation of GATA-4 is abolished by MEK inhibitors or the mutation of the ERK phosphorylation site (S105A), suggesting that HGF activates GATA-4 via MEK-ERK pathway-dependent phosphorylation. HGF enhances the expression of anti-apoptotic Bcl-x(L), and this is blocked by dominant negative mutants of MEK or GATA-4. Forced expression of wild-type GATA-4, but not the GATA-4 mutant (S105A) increases the expression of Bcl-x(L). Furthermore, expression of the GATA-4 mutant (S105A) suppresses HGF-mediated protection of cells against daunorubicin-induced apoptosis. These results demonstrate that HGF protects cardiac muscle cells against apoptosis via a signaling pathway involving MEK/ERK-dependent phosphorylation of GATA-4.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fator de Transcrição GATA4 , Fator de Crescimento de Hepatócito/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Miócitos Cardíacos/citologia , Fosforilação , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais
16.
Mol Cell Biol ; 22(5): 1317-28, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11839799

RESUMO

We identified cDNAs encoding the Xenopus Smad proteins most closely related to mammalian Smad8, and we present a functional analysis of this activity (also referred to recently as xSmad11). Misexpression experiments indicate that xSmad8(11) regulates pathways distinct from those regulated by the closely related xSmad1. Embryos that develop from eggs depleted of xSmad8(11) mRNA fail to gastrulate; instead, at the time of gastrulation, they initiate a widespread program of apoptosis, via a CPP32/caspase 3 pathway. Embryos that avoid this fate display gastrulation defects. Activation of apoptosis is rescued by expression of xSmad8(11) but not xSmad1. Our results demonstrate an embryonic requirement for Smad8(11) activity and show that a maternally derived Smad signaling pathway is required for gastrulation and for mediating a cell survival program during early embryogenesis. We suggest that xSmad8(11) functions as part of a maternally derived mechanism shown previously by others to monitor Xenopus early embryonic cell cycles.


Assuntos
Apoptose , Proteínas de Ligação a DNA/metabolismo , Gástrula/fisiologia , RNA Mensageiro Estocado/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Xenopus , Xenopus laevis/embriologia , Sequência de Aminoácidos , Animais , Padronização Corporal , Proteínas Morfogenéticas Ósseas/genética , Sequência Conservada , DNA Complementar , Proteínas de Ligação a DNA/genética , Ectoderma , Regulação da Expressão Gênica no Desenvolvimento , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas Smad , Proteína Smad8 , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...