Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Biol ; 461(2): 124-131, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32035085

RESUMO

Development of multi-chambered heart is associated with spatio-temporal regulation of gene expression. A basic helix-loop-helix transcription factor Hey2 is specifically expressed in the embryonic mouse ventricles and is indispensable for ventricular myocyte differentiation, compartment identity and morphogenesis of the heart. However, how Hey2 transcription is precisely regulated in the heart remains unclear. In this study, we identified a distal Hey2 enhancer conserved in the mouse and human to possess specific transcriptional activity in ventricular free wall myocytes at the looping stage of cardiac development. Deletion of the enhancer significantly decreased endogenous Hey2 expression in the ventricular myocardium but not in other tissues of mouse embryos. Mutation/deletion of the conserved binding sites for T-box and Gata proteins, but not NK-2 proteins, abolished the enhancer activity, and Tbx20 null mice completely lost the enhancer activity in the embryonic ventricles. Luciferase reporter analysis suggested that the ventricular enhancer activity was controlled by Tbx20 through its DNA binding and cooperative function with cardiac Gata proteins. These results delineate a regulatory mechanism of ventricular Hey2 expression and help fully understand molecular cascades in myocardial cell differentiation and cardiac morphogenesis during embryonic development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Elementos Facilitadores Genéticos , Fator de Transcrição GATA4/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Ventrículos do Coração/embriologia , Proteínas Repressoras/biossíntese , Proteínas com Domínio T/fisiologia , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sequência Conservada , Genes Reporter , Ventrículos do Coração/metabolismo , Humanos , Mamíferos/genética , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/genética , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie
2.
Mediators Inflamm ; 2021: 3698386, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34545275

RESUMO

Coronary artery disease (CAD) has been the leading cause of morbidity and mortality worldwide, and its pathogenesis is closely related with the proliferation and migration of vascular smooth muscle cell (VSMC). We previously reported a truncated GATA4 protein lacking C-terminus induced by p.S335X mutation in cardiomyocyte from ventricular septal defect (VSD) patients. However, it is still unclear whether GATA4 p.S335X mutation could influence the development of CAD. GATA4 wild-type (WT) and p.S335X mutant (MU) overexpression plasmids were constructed and transfected transiently into rat coronary artery smooth muscle cell (RCSMC) to observe the proliferative and migratory abilities by MTS and wound healing assay, respectively. PCR array was used to preliminarily detect the expression of phenotypic modulation-related genes, and QRT-PCR was then carried out to verify the screened differentially expressed genes (DEGs). The results showed that, when stimulated by fetal bovine serum (10%) for 24 h or tumor necrosis factor-α (10 or 30 ng/ml) for 10 or 24 h, deletion of GATA4 C-terminus by p.S335X mutation in GATA4 enhanced the proliferation of RCSMC, without alteration of the migration capability. Twelve DEGs, including Fas, Hbegf, Itga5, Aimp1, Cxcl1, Il15, Il2rg, Il7, Tnfsf10, Il1r1, Irak1, and Tlr3, were screened and identified as phenotypic modulation-related genes. Our data might be beneficial for further exploration regarding the mechanisms of GATA4 p.S335X mutation on the phenotypic modulation of coronary VSMC.


Assuntos
Vasos Coronários/fisiologia , Fator de Transcrição GATA4/genética , Músculo Liso Vascular/citologia , Mutação , Miócitos de Músculo Liso/fisiologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Doença da Artéria Coronariana/etiologia , Fator de Transcrição GATA4/fisiologia , Músculo Liso Vascular/fisiologia , Fenótipo , Ratos
3.
Circ Res ; 122(8): 1069-1083, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29475983

RESUMO

RATIONALE: Multilineage-differentiating stress enduring (Muse) cells, pluripotent marker stage-specific embryonic antigen-3+ cells, are nontumorigenic endogenous pluripotent-like stem cells obtainable from various tissues including the bone marrow. Their therapeutic efficiency has not been validated in acute myocardial infarction. OBJECTIVE: The main objective of this study is to clarify the efficiency of intravenously infused rabbit autograft, allograft, and xenograft (human) bone marrow-Muse cells in a rabbit acute myocardial infarction model and their mechanisms of tissue repair. METHODS AND RESULTS: In vivo dynamics of Nano-lantern-labeled Muse cells showed preferential homing of the cells to the postinfarct heart at 3 days and 2 weeks, with ≈14.5% of injected GFP (green fluorescent protein)-Muse cells estimated to be engrafted into the heart at 3 days. The migration and homing of the Muse cells was confirmed pharmacologically (S1PR2 [sphingosine monophosphate receptor 2]-specific antagonist JTE-013 coinjection) and genetically (S1PR2-siRNA [small interfering ribonucleic acid]-introduced Muse cells) to be mediated through the S1P (sphingosine monophosphate)-S1PR2 axis. They spontaneously differentiated into cells positive for cardiac markers, such as cardiac troponin-I, sarcomeric α-actinin, and connexin-43, and vascular markers. GCaMP3 (GFP-based Ca calmodulin probe)-labeled Muse cells that engrafted into the ischemic region exhibited increased GCaMP3 fluorescence during systole and decreased fluorescence during diastole. Infarct size was reduced by ≈52%, and the ejection fraction was increased by ≈38% compared with vehicle injection at 2 months, ≈2.5 and ≈2.1 times higher, respectively, than that induced by mesenchymal stem cells. These effects were partially attenuated by the administration of GATA4-gene-silenced Muse cells. Muse cell allografts and xenografts efficiently engrafted and recovered functions, and allografts remained in the tissue and sustained functional recovery for up to 6 months without immunosuppression. CONCLUSIONS: Muse cells may provide reparative effects and robust functional recovery and may, thus, provide a novel strategy for the treatment of acute myocardial infarction.


Assuntos
Lisofosfolipídeos/fisiologia , Infarto do Miocárdio/cirurgia , Células-Tronco Pluripotentes/transplante , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Aloenxertos , Animais , Autoenxertos , Diferenciação Celular , Movimento Celular/fisiologia , Fator de Transcrição GATA4/antagonistas & inibidores , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/fisiologia , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/análise , Xenoenxertos , Humanos , Luciferases/análise , Proteínas Luminescentes/análise , Masculino , Infarto do Miocárdio/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Coelhos , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes de Fusão/análise , Especificidade da Espécie , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato
4.
Development ; 143(5): 780-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26932670

RESUMO

GATA4 and GATA6 are zinc finger transcription factors that have important functions in several mesodermal and endodermal organs, including heart, liver and pancreas. In humans, heterozygous mutations of either factor are associated with pancreatic agenesis; however, homozygous deletion of both Gata4 and Gata6 is necessary to disrupt pancreas development in mice. In this study, we demonstrate that arrested pancreatic development in Gata4(fl/fl); Gata6(fl/fl); Pdx1:Cre (pDKO) embryos is accompanied by the transition of ventral and dorsal pancreatic fates into intestinal or stomach lineages, respectively. These results indicate that GATA4 and GATA6 play essential roles in maintaining pancreas identity by regulating foregut endodermal fates. Remarkably, pancreatic anlagen derived from pDKO embryos also display a dramatic upregulation of hedgehog pathway components, which are normally absent from the presumptive pancreatic endoderm. Consistent with the erroneous activation of hedgehog signaling, we demonstrate that GATA4 and GATA6 are able to repress transcription through the sonic hedgehog (Shh) endoderm-specific enhancer MACS1 and that GATA-binding sites within this enhancer are necessary for this repressive activity. These studies establish the importance of GATA4/6-mediated inhibition of hedgehog signaling as a major mechanism regulating pancreatic endoderm specification during patterning of the gut tube.


Assuntos
Endoderma/fisiologia , Fator de Transcrição GATA4/fisiologia , Fator de Transcrição GATA6/fisiologia , Pâncreas/embriologia , Animais , Sequência de Bases , Padronização Corporal , Linhagem da Célula , Imunoprecipitação da Cromatina , Coenzima A Ligases/fisiologia , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Heterozigoto , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/fisiologia , Mutação , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Transdução de Sinais
5.
J Gastroenterol Hepatol ; 34(12): 2196-2205, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30995348

RESUMO

BACKGROUND AND AIM: The high mortality and poor prognosis of hepatocellular carcinoma (HCC) have raised the public attention. Gene therapy is considered as a promising treatment option for cancer; thus, finding a new therapeutic target for HCC is urgently needed. GATA4 is a tumor suppressor gene in multiple cancers, but its role in HCC is unclear. In this study, we explored the function of GATA4 in HCC. METHODS: Reverse transcription-polymerase chain reaction and quantitative polymerase chain reaction were used to detect the mRNA expression of GATA4 in HCC cells and tissues. Cell viability, transwell, colony formation, and flow cytometry assays were applied to examine different aspects of biological effects of GATA4 in vitro. Xenografts, immunohistochemistry, and terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assays were performed to evaluate the effect of GATA4 on tumorigenicity in vivo. Western blotting, immunofluorescence, and ß-galactosidase staining were used to investigate the mechanism underlying the function of GATA4. RESULTS: We found that GATA4 was silenced in 15/19 (79%) HCC tissues. Restoring the expression of GATA4 induced G0 /G1 phase arrest, promoted apoptosis, suppressed HCC proliferation in vitro, and inhibited HCC tumor growth in vivo. Our data further showed that the ectopic expression of GATA4 induced cellular senescence through regulating nuclear factor-κB and inducing mesenchymal-to-epithelial transition. CONCLUSIONS: Our data demonstrated that by inducing cellular senescence and mesenchymal-to-epithelial transition, GATA4 plays a crucial role as a tumor suppressor in HCC. It may thus be a potential cancer therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Senescência Celular/fisiologia , Fator de Transcrição GATA4/fisiologia , Neoplasias Hepáticas/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Senescência Celular/genética , Regulação para Baixo/fisiologia , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Fator de Transcrição GATA4/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica , Xenoenxertos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos Nus , NF-kappa B/fisiologia , Invasividade Neoplásica , Transplante de Neoplasias , RNA Mensageiro/genética , RNA Neoplásico/genética , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
6.
Reproduction ; 156(4): 343­351, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30306767

RESUMO

Reduced contractility of the testicular peritubular myoid (PTM) cells may contribute to human male subfertility or infertility. Transcription factor GATA4 in Sertoli and Leydig cells is essential for murine spermatogenesis, but limited attention has been paid to the potential role of GATA4 in PTM cells. In primary cultures of mouse PTM cells, siRNA knockdown of GATA4 increased the contractile activity, while GATA4 overexpression significantly attenuated the contractility of PTM cells using a collagen gel contraction assay. Using RNA sequencing and qRT-PCR, we identified a set of genes that exhibited opposite expressional alternation between Gata4 siRNA vs nontargeting siRNA-treated PTM cells and Gata4 adenovirus vs control adenovirus-treated PTM cells. Notably, ion channels, smooth muscle function, cytokines and chemokines, cytoskeleton, adhesion and extracellular matrix were the top four enriched pathways, as revealed by cluster analysis. Natriuretic peptide type B (NPPB) content was significantly upregulated by GATA4 overexpression in both PTM cells and their culture supernatant. More importantly, the addition of 100 µM NPPB could abolish the promoting effect of Gata4 silencing on PTM cell contraction. Taken together, we suggest that the inhibitory action of GATA4 on PTM cell contraction is mediated at least partly by regulating genes belonging to smooth muscle contraction pathway (e.g. Nppb).


Assuntos
Fator de Transcrição GATA4/fisiologia , Testículo/fisiologia , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Peptídeo Natriurético Encefálico/metabolismo , Cultura Primária de Células , Espermatogênese , Testículo/citologia
7.
Dev Biol ; 410(1): 24-35, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26719127

RESUMO

Adult intestinal tissues, exposed to the external environment, play important roles including barrier and nutrient-absorption functions. These functions are ensured by adequately controlled rapid-cell metabolism. GATA transcription factors play essential roles in the development and maintenance of adult intestinal tissues both in vertebrates and invertebrates. We investigated the roles of GATAe, the Drosophila intestinal GATA factor, in adult midgut homeostasis with its first-generated knock-out mutant as well as cell type-specific RNAi and overexpression experiments. Our results indicate that GATAe is essential for proliferation and maintenance of intestinal stem cells (ISCs). Also, GATAe is involved in the differentiation of enterocyte (EC) and enteroendocrine (ee) cells in both Notch (N)-dependent and -independent manner. The results also indicate that GATAe has pivotal roles in maintaining normal epithelial homeostasis of the Drosophila adult midgut through interaction of N signaling. Since recent reports showed that mammalian GATA-6 regulates normal and cancer stem cells in the adult intestinal tract, our data also provide information on the evolutionally conserved roles of GATA factors in stem-cell regulation.


Assuntos
Diferenciação Celular , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Fatores de Transcrição GATA/fisiologia , Intestinos/citologia , Células-Tronco/citologia , Envelhecimento , Animais , Drosophila melanogaster/citologia , Fator de Transcrição GATA4/fisiologia , Fator de Transcrição GATA6/fisiologia
8.
Dev Biol ; 410(2): 213-222, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26687508

RESUMO

Gata4 and Gata6 are closely related transcription factors that are essential for the development of a number of embryonic tissues. While they have nearly identical DNA-binding domains and similar patterns of expression, Gata4 and Gata6 null embryos have strikingly different embryonic lethal phenotypes. To determine whether the lack of redundancy is due to differences in protein function or Gata4 and Gata6 expression domains, we generated mice that contained the Gata6 cDNA in place of the Gata4 genomic locus. Gata4(Gata6/Gata6) embryos survived through embryonic day (E)12.5 and successfully underwent ventral folding morphogenesis, demonstrating that Gata6 is able to replace Gata4 function in extraembryonic tissues. Surprisingly, Gata6 is unable to replace Gata4 function in the septum transversum mesenchyme or the epicardium, leading to liver agenesis and lethal heart defects in Gata4(Gata6/Gata6) embryos. These studies suggest that Gata4 has evolved distinct functions in the development of these tissues that cannot be performed by Gata6, even when it is provided in the identical expression domain. Our work has important implications for the respective mechanisms of Gata function during development, as well as the functional evolution of these essential transcription factors.


Assuntos
Fator de Transcrição GATA4/fisiologia , Coração/embriologia , Fígado/embriologia , Animais , DNA Complementar/genética , Fator de Transcrição GATA4/genética , Células HEK293 , Humanos , Camundongos
9.
Reproduction ; 153(2): 211-220, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27864336

RESUMO

Generation of male germ cells from pluripotent cells could provide male gametes for treating male infertility and offer an ideal model for unveiling molecular mechanisms of spermatogenesis. However, the influence and exact molecular mechanisms, especially downstream effectors of BMP4 signaling pathways, in male germ cell differentiation of the induce pluripotent stem (iPS) cells, remain unknown. This study was designed to explore the role and mechanism of BMP4 signaling in the differentiation of mouse iPS cells to male germ cells. Embryoid body (EB) formation and recombinant BMP4 or Noggin were utilized to evaluate the effect of BMP4 on male germ cell generation from mouse iPS cells. Germ cell-specific genes and proteins as well as the downstream effectors of BMP4 signaling pathway were assessed using real-time PCR and Western blots. We found that BMP4 ligand and its multiple receptors, including BMPR1a, BMPR1b and BMPR2, were expressed in mouse iPS cells. Real-time PCR and Western blots revealed that BMP4 could upregulate the levels of genes and proteins for germ cell markers in iPS cells-derived EBs, whereas Noggin decreased their expression in these cells. Moreover, Smad1/5 phosphorylation, Gata4 transcription and the transcripts of Id1 and Id2 were enhanced by BMP4 but decreased when exposed to Noggin. Collectively, these results suggest that BMP4 promotes the generation of male germ cells from iPS cells via Smad1/5 pathway and the activation of Gata4, Id1 and Id2 This study thus offers novel insights into molecular mechanisms underlying male germ cell development.


Assuntos
Proteína Morfogenética Óssea 4/fisiologia , Diferenciação Celular/fisiologia , Células Germinativas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Western Blotting , Proteína Morfogenética Óssea 4/genética , Linhagem Celular , Fator de Transcrição GATA4/fisiologia , Expressão Gênica , Células-Tronco Pluripotentes Induzidas/fisiologia , Proteína 1 Inibidora de Diferenciação/fisiologia , Proteína 2 Inibidora de Diferenciação/fisiologia , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Espermatozoides/citologia
10.
Proc Natl Acad Sci U S A ; 111(4): 1415-20, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24474767

RESUMO

The G1 cyclins play a pivotal role in regulation of cell differentiation and proliferation. The mechanisms underlying their cell-specific roles are incompletely understood. Here, we show that a G1 cyclin, cyclin D2 (CycD2), enhances the activity of transcription factor GATA4, a key regulator of cardiomyocyte growth and differentiation. GATA4 recruits CycD2 to its target promoters, and their interaction results in synergistic activation of GATA-dependent transcription. This effect is specific to CycD2 because CycD1 is unable to potentiate activity of GATA4 and is CDK-independent. GATA4 physically interacts with CycD2 through a discreet N-terminal activation domain that is essential for the cardiogenic activity of GATA4. Human mutations in this domain that are linked to congenital heart disease interfere with CycD2-GATA4 synergy. Cardiogenesis assays in Xenopus embryos indicate that CycD2 enhances the cardiogenic function of GATA4. Together, our data uncover a role for CycD2 as a cardiogenic coactivator of GATA4 and suggest a paradigm for cell-specific effects of cyclin Ds.


Assuntos
Ciclina D2/fisiologia , Fator de Transcrição GATA4/fisiologia , Coração/embriologia , Organogênese/fisiologia , Sequência de Aminoácidos , Animais , Ciclina D2/química , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Transativadores/fisiologia
11.
Biochim Biophys Acta ; 1849(12): 1411-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26477491

RESUMO

Intestinal epithelial cells are exposed to luminal bacterial threat and require adequate defense mechanisms to ensure host protection and epithelium regeneration against possible deleterious damage. Differentiated intestinal epithelial cells produce antimicrobial and regenerative components that protect against such challenges. Few intestinal specific transcription factors have been identified to control the switching from repression to activation of this class of gene. Herein, we show that gene transcription of some regenerating islet-derived (REG) family members is dependent on the transcription factor GATA-4. Silencing of GATA-4 expression in cultured intestinal epithelial cells identified Reg3ß as a target gene using an unbiased approach of gene expression profiling. Co-transfection and RNA interference assays identified complex GATA-4-interactive transcriptional components required for the activation or repression of Reg3ß gene activity. Conditional deletion of Gata4 in the mouse intestinal epithelium supported its regulatory role for Reg1, Reg3α, Reg3ß and Reg3γ genes. Reg1 dramatic down-modulation of expression in Gata4 conditional null mice was associated with a significant decrease in intestinal epithelial cell migration. Altogether, these results identify a novel and complex role for GATA-4 in the regulation of REG family members gene expression.


Assuntos
Células Epiteliais/metabolismo , Fator de Transcrição GATA4/fisiologia , Regulação da Expressão Gênica/genética , Mucosa Intestinal/citologia , Família Multigênica , Transcrição Gênica , Animais , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Fator de Transcrição CDX2 , Diferenciação Celular/genética , Linhagem Celular , Técnicas de Cocultura , Fator de Transcrição GATA4/classificação , Fator de Transcrição GATA4/deficiência , Fator de Transcrição GATA4/genética , Genes Reporter , Proteínas de Homeodomínio/metabolismo , Lectinas Tipo C/metabolismo , Litostatina/metabolismo , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas Associadas a Pancreatite , Estrutura Terciária de Proteína , Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
12.
Hum Reprod ; 31(6): 1300-14, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27083540

RESUMO

STUDY QUESTION: What are the functional characteristics and transcriptional regulators of human trophoblast progenitor cells (TBPCs)? SUMMARY ANSWER: TBPC lines established from the human smooth chorion by cell sorting for integrin α4 expressed markers of stemness and trophoblast (TB) stage-specific antigens, invaded Matrigel substrates and contributed to the cytotrophoblasts (CTBs) layer of smooth chorion explants with high-mobility group protein HMGI-C (HMGA2) and transcription factor GATA-4 (GATA4) controlling their progenitor state and TB identity. WHAT IS KNOWN ALREADY: Previously, we reported the derivation of TBPC lines by trypsinization of colonies that formed in cultures of chorionic mesenchyme cells that were treated with an activin nodal inhibitor. Microarray analyses showed that, among integrins, α4 was most highly expressed, and identified HMGA2 and GATA4 as potential transcriptional regulators. STUDY DESIGN, SIZE, DURATION: The aim of this study was to streamline TBPC derivation across gestation. High-cell surface expression of integrin α4 enabled the use of a fluorescence-activated cell sorter (FACS) approach for TBPC isolation from the human smooth chorion (n = 6 lines). To confirm their TBPC identity, we profiled their expression of stemness and TB markers, and growth factor receptors. At a functional level, we assayed their invasive capacity (n = 3) and tropism for the CTB layer of the smooth chorion (n = 3). At a molecular level, we studied the roles of HMGA2 and GATA4. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: Cells were enzymatically disassociated from the human smooth chorion across gestation. FACS was used to isolate the integrin α4-positive population. In total, we established six TBPC lines, two per trimester. Their identity was determined by immunolocalization of a suite of antigens. Function was assessed via Matrigel invasion and co-culture with explants of the human smooth chorion. An siRNA approach was used to down-regulate HMGA2 and GATA4 expression and the results were confirmed by immunoblotting and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analyses. The endpoints analyzed included proliferation, as determined by 5-bromo-2'-deoxyuridine (BrDU) incorporation, and the expression of stage-specific antigens and hormones, as determined by qRT-PCR and immunostaining approaches. MAIN RESULTS AND THE ROLE OF CHANCE: As with the original cell lines, the progenitors expressed a combination of human embryonic stem cell and TB markers. Upon differentiation, they primarily formed CTBs, which were capable of Matrigel invasion. Co-culture of the cells with smooth chorion explants enabled their migration through the mesenchyme after which they intercalated within the chorionic CTB layer. Down-regulation of HMGA2 showed that this DNA-binding protein governed their self-renewal. Both HMGA2 and GATA4 had pleitropic effects on the cells' progenitor state and TB identity. LIMITATIONS, REASONS FOR CAUTION: This study supported our hypothesis that TBPCs from the chorionic mesenchyme can contribute to the subpopulation of CTBs that reside in the smooth chorion. In the absence of in vivo data, which is difficult to obtain in humans, the results have the limitations common to all in vitro studies. WIDER IMPLICATIONS OF THE FINDINGS: The accepted view is that progenitors reside among the villous CTB subpopulation. Here, we show that TBPCs also reside in the mesenchymal layer of the smooth chorion throughout gestation. We theorize that they can contribute to the CTB layer in this region. This phenomenon may be particularly important in pathological situations when CTBs of the smooth chorion might provide a functional reserve for CTBs of the placenta proper. STUDY FUNDING/COMPETING INTERESTS: Research reported in this publication was supported by the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health under award P50HD055764. O.G., N.L., K.O., A.P., T.G.-G., M.K., A.B., M.G. have nothing to disclose. S.J.F. received licensing fees and royalties from SeraCare Life Sciences for trisomic TBPC lines that were derived according to the methods described in this manuscript. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Fator de Transcrição GATA4/fisiologia , Integrina alfa4/metabolismo , Trofoblastos/metabolismo , Diferenciação Celular , Linhagem Celular , Córion/citologia , Córion/metabolismo , Técnicas de Cocultura , Citometria de Fluxo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Proteína HMGA2/fisiologia , Humanos , Integrina alfa4/genética , Elementos Reguladores de Transcrição
13.
PLoS Genet ; 9(7): e1003629, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874227

RESUMO

In mammals, both testis and ovary arise from a sexually undifferentiated precursor, the genital ridge, which first appears during mid-gestation as a thickening of the coelomic epithelium on the ventromedial surface of the mesonephros. At least four genes (Lhx9, Sf1, Wt1, and Emx2) have been demonstrated to be required for subsequent growth and maintenance of the genital ridge. However, no gene has been shown to be required for the initial thickening of the coelomic epithelium during genital ridge formation. We report that the transcription factor GATA4 is expressed in the coelomic epithelium of the genital ridge, progressing in an anterior-to-posterior (A-P) direction, immediately preceding an A-P wave of epithelial thickening. Mouse embryos conditionally deficient in Gata4 show no signs of gonadal initiation, as their coelomic epithelium remains a morphologically undifferentiated monolayer. The failure of genital ridge formation in Gata4-deficient embryos is corroborated by the absence of the early gonadal markers LHX9 and SF1. Our data indicate that GATA4 is required to initiate formation of the genital ridge in both XX and XY fetuses, prior to its previously reported role in testicular differentiation of the XY gonad.


Assuntos
Diferenciação Celular , Fator de Transcrição GATA4/genética , Genitália Feminina/crescimento & desenvolvimento , Genitália Masculina/crescimento & desenvolvimento , Processos de Determinação Sexual/genética , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário , Feminino , Fator de Transcrição GATA4/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/citologia , Gônadas/crescimento & desenvolvimento , Masculino , Camundongos , Ovário/crescimento & desenvolvimento , Testículo/crescimento & desenvolvimento
14.
Dev Biol ; 386(1): 204-15, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24380800

RESUMO

During early development, GATA factors have been shown to be important for key events of coronary vasculogenesis, including formation of the epicardium. Myocardial GATA factors are required for coronary vascular (CV) formation; however, the role of epicardial localized GATAs in this process has not been addressed. The current study was conducted to investigate the molecular mechanisms by which the epicardium controls coronary vasculogenesis, focusing on the role of epicardial GATAs in establishing the endothelial plexus during early coronary vasculogenesis. To address the role of epicardial GATAs, we ablated GATA4 and GATA6 transcription factors specifically from the mouse epicardium and found that the number of endothelial cells in the sub-epicardium was drastically reduced, and concomitant coronary vascular plexus formation was significantly compromised. Here we present evidence for a novel role for epicardial GATA factors in controlling plexus formation by recruiting endothelial cells to the sub-epicardium.


Assuntos
Vasos Coronários/metabolismo , Endotélio Vascular/metabolismo , Fator de Transcrição GATA4/fisiologia , Fator de Transcrição GATA6/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Pericárdio/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Cruzamentos Genéticos , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA6/genética , Genótipo , Coração/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Transdução de Sinais , Fatores de Tempo
15.
Biochim Biophys Acta ; 1840(6): 2060-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24582939

RESUMO

BACKGROUND: Transplanted mesenchymal stem cells (MSC) can differentiate into cardiac cells that have the potential to contribute to heart repair following ischemic injury. Overexpression of GATA-4 can significantly increase differentiation of MSC into cardiomyocytes (CM). However, the specific impact of GATA-4 overexpression on the electrophysiological properties of MSC-derived CM has not been well documented. METHODS: Adult rat bone marrow MSC were retrovirally transduced with GATA-4 (MSC(GATA-4)) and GFP (MSC(Null)) and subsequently co-cultured with neonatal rat ventricular cardiomyocytes (CM). Electrophysiological properties and mRNA levels of ion channels were assessed in MSC using patch-clamp technology and real-time PCR. RESULTS: MSC(GATA-4) exhibited higher levels of the TTX-sensitive Na(+) current (INa.TTX), L-type calcium current (ICa.L), transient outward K(+) current (Ito), delayed rectifier K(+) current (IKDR) and inwardly rectifying K(+) current (IK1) channel activities reflective of electrophysiological characteristics of CM. Real-time PCR analyses showed that MSC(GATA-4) exhibited upregulated mRNA levels of Kv1.2, Kv2.1, SCN2a1, CCHL2a, KV1.4 and Kir1.1 channels versus MSC(Null). Interestingly, MSC(GATA-4) treated with IGF-1 neutralizing antibodies resulted in a significant decrease in Kir1.1, Kv2.1, KV1.4, CCHL2a and SCN2a1 channel mRNA expression. Similarly, MSC(GATA-4) treated with VEGF neutralizing antibodies also resulted in an attenuated expression of Kv2.1, Kv1.2, Kv1.4, Kir1.1, CCHL2a and SCN2a1 channel mRNAs. CONCLUSIONS: GATA-4 overexpression increases Ito, IKDR, IK1, INa.TTX and ICa.L currents in MSC. Cytokine (VGEF and IGF-1) release from GATA-4 overexpressing MSC can partially account for the upregulated ion channel mRNA expression. GENERAL SIGNIFICANCE: Our results highlight the ability of GATA4 to boost the cardiac electrophysiological potential of MSC.


Assuntos
Fator de Transcrição GATA4/fisiologia , Células-Tronco Mesenquimais/fisiologia , Animais , Células Cultivadas , Citocinas/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/fisiologia
16.
Biochim Biophys Acta ; 1839(11): 1273-82, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24878542

RESUMO

GATA4 is expressed in the proximal 85% of small intestine where it promotes a proximal intestinal ('jejunal') identity while repressing a distal intestinal ('ileal') identity, but its molecular mechanisms are unclear. Here, we tested the hypothesis that GATA4 promotes a jejunal versus ileal identity in mouse intestine by directly activating and repressing specific subsets of absorptive enterocyte genes by modulating the acetylation of histone H3, lysine 27 (H3K27), a mark of active chromatin, at sites of GATA4 occupancy. Global analysis of mouse jejunal epithelium showed a statistically significant association of GATA4 occupancy with GATA4-regulated genes. Occupancy was equally distributed between down- and up-regulated targets, and occupancy sites showed a dichotomy of unique motif over-representation at down- versus up-regulated genes. H3K27ac enrichment at GATA4-binding loci that mapped to down-regulated genes (activation targets) was elevated, changed little upon conditional Gata4 deletion, and was similar to control ileum, whereas H3K27ac enrichment at GATA4-binding loci that mapped to up-regulated genes (repression targets) was depleted, increased upon conditional Gata4 deletion, and approached H3K27ac enrichment in wild-type control ileum. These data support the hypothesis that GATA4 both activates and represses intestinal genes, and show that GATA4 represses an ileal program of gene expression in the proximal small intestine by inhibiting the acetylation of H3K27.


Assuntos
Fator de Transcrição GATA4/fisiologia , Histona Acetiltransferases/antagonistas & inibidores , Histonas/metabolismo , Íleo/metabolismo , Acetilação , Animais , Células Cultivadas , Regulação para Baixo/genética , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Intestino Delgado/metabolismo , Lisina/metabolismo , Camundongos , Camundongos Transgênicos
17.
J Hepatol ; 63(3): 697-704, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26022694

RESUMO

BACKGROUND & AIMS: Regulation of bile acid homeostasis in mammals is a complex process regulated via extensive cross-talk between liver, intestine and intestinal microbiota. Here we studied the effects of gut microbiota on bile acid homeostasis in mice. METHODS: Bile acid homeostasis was assessed in four mouse models. Germfree mice, conventionally-raised mice, Asbt-KO mice and intestinal-specific Gata4-iKO mice were treated with antibiotics (bacitracin, neomycin and vancomycin; 100 mg/kg) for five days and subsequently compared with untreated mice. RESULTS: Attenuation of the bacterial flora by antibiotics strongly reduced fecal excretion and synthesis of bile acids, but increased the expression of the bile acid synthesis enzyme CYP7A1. Similar effects were seen in germfree mice. Intestinal bile acid absorption was increased and accompanied by increases in plasma bile acid levels, biliary bile acid secretion and enterohepatic cycling of bile acids. In the absence of microbiota, the expression of the intestinal bile salt transporter Asbt was strongly increased in the ileum and was also expressed in more proximal parts of the small intestine. Most of the effects of antibiotic treatment on bile acid homeostasis could be prevented by genetic inactivation of either Asbt or the transcription factor Gata4. CONCLUSIONS: Attenuation of gut microbiota alters Gata4-controlled expression of Asbt, increasing absorption and decreasing synthesis of bile acids. Our data support the concept that under physiological conditions microbiota stimulate Gata4, which suppresses Asbt expression, limiting the expression of this transporter to the terminal ileum. Our studies expand current knowledge on the bacterial control of bile acid homeostasis.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fator de Transcrição GATA4/fisiologia , Microbioma Gastrointestinal/fisiologia , Absorção Intestinal , Transportadores de Ânions Orgânicos Dependentes de Sódio/fisiologia , Simportadores/fisiologia , Animais , Antibacterianos/farmacologia , Colesterol 7-alfa-Hidroxilase/genética , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise
18.
Hepatology ; 59(6): 2358-70, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24415412

RESUMO

UNLABELLED: The zinc finger transcription factor GATA4 controls specification and differentiation of multiple cell types during embryonic development. In mouse embryonic liver, Gata4 is expressed in the endodermal hepatic bud and in the adjacent mesenchyme of the septum transversum. Previous studies have shown that Gata4 inactivation impairs liver formation. However, whether these defects are caused by loss of Gata4 in the hepatic endoderm or in the septum transversum mesenchyme remains to be determined. In this study, we have investigated the role of mesenchymal GATA4 activity in liver formation. We have conditionally inactivated Gata4 in the septum transversum mesenchyme and its derivatives by using Cre/loxP technology. We have generated a mouse transgenic Cre line, in which expression of Cre recombinase is controlled by a previously identified distal Gata4 enhancer. Conditional inactivation of Gata4 in hepatic mesenchymal cells led to embryonic lethality around mouse embryonic stage 13.5, likely as a consequence of fetal anemia. Gata4 knockout fetal livers exhibited reduced size, advanced fibrosis, accumulation of extracellular matrix components and hepatic stellate cell (HSC) activation. Haploinsufficiency of Gata4 accelerated CCl4 -induced liver fibrosis in adult mice. Moreover, Gata4 expression was dramatically reduced in advanced hepatic fibrosis and cirrhosis in humans. CONCLUSIONS: Our data demonstrate that mesenchymal GATA4 activity regulates HSC activation and inhibits the liver fibrogenic process.


Assuntos
Regulação para Baixo , Fator de Transcrição GATA4/fisiologia , Cirrose Hepática/metabolismo , Fígado/embriologia , Animais , Intoxicação por Tetracloreto de Carbono/complicações , Linhagem Celular , Matriz Extracelular/metabolismo , Células Estreladas do Fígado/fisiologia , Humanos , Integrases , Cirrose Hepática/etiologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo
19.
Arterioscler Thromb Vasc Biol ; 34(6): 1260-71, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24675663

RESUMO

OBJECTIVE: Right ventricular (RV) failure is the leading cause of death in various cardiopulmonary diseases, including pulmonary hypertension. It is generally considered that the RV is vulnerable to pressure overload as compared with the left ventricle (LV). However, as compared with LV failure, the molecular mechanisms of RV failure are poorly understood, and hence therapeutic targets of the disorder remain to be elucidated. Thus, we aimed to identify molecular therapeutic targets for RV failure in a mouse model of pressure overload. APPROACH AND RESULTS: To induce pressure overload to respective ventricles, we performed pulmonary artery constriction or transverse aortic constriction in mice. We first performed microarray analysis and found that the molecules related to RhoA/Rho-kinase and integrin pathways were significantly upregulated in the RV with pulmonary artery constriction compared with the LV with transverse aortic constriction. Then, we examined the responses of both ventricles to chronic pressure overload in vivo. We demonstrated that compared with transverse aortic constriction, pulmonary artery constriction caused greater extents of mortality, Rho-kinase expression (especially ROCK2 isoform), and oxidative stress in pressure-overloaded RV, reflecting the weakness of the RV in response to pressure overload. Furthermore, mice with myocardial-specific overexpression of dominant-negative Rho-kinase showed resistance to pressure overload-induced hypertrophy and dysfunction associated with reduced oxidative stress. Finally, dominant-negative Rho-kinase mice showed a significantly improved long-term survival in both pulmonary artery constriction and transverse aortic constriction as compared with littermate controls. CONCLUSION: These results indicate that the Rho-kinase pathway plays a crucial role in RV hypertrophy and dysfunction, suggesting that the pathway is a novel therapeutic target of RV failure in humans.


Assuntos
Hipertrofia Ventricular Direita/etiologia , Disfunção Ventricular Direita/etiologia , Quinases Associadas a rho/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fibrose , Fator de Transcrição GATA4/fisiologia , Hipertensão Pulmonar/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Transdução de Sinais/fisiologia
20.
Circ Res ; 111(9): 1147-56, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22931955

RESUMO

RATIONALE: After myocardial infarction (MI), massive cell death in the myocardium initiates fibrosis and scar formation, leading to heart failure. We recently found that a combination of 3 cardiac transcription factors, Gata4, Mef2c, and Tbx5 (GMT), reprograms fibroblasts directly into functional cardiomyocytes in vitro. OBJECTIVE: To investigate whether viral gene transfer of GMT into infarcted hearts induces cardiomyocyte generation. METHODS AND RESULTS: Coronary artery ligation was used to generate MI in the mouse. In vitro transduction of GMT retrovirus converted cardiac fibroblasts from the infarct region into cardiomyocyte-like cells with cardiac-specific gene expression and sarcomeric structures. Injection of the green fluorescent protein (GFP) retrovirus into mouse hearts, immediately after MI, infected only proliferating noncardiomyocytes, mainly fibroblasts, in the infarct region. The GFP expression diminished after 2 weeks in immunocompetent mice but remained stable for 3 months in immunosuppressed mice, in which cardiac induction did not occur. In contrast, injection of GMT retrovirus into α-myosin heavy chain (αMHC)-GFP transgenic mouse hearts induced the expression of αMHC-GFP, a marker of cardiomyocytes, in 3% of virus-infected cells after 1 week. A pooled GMT injection into the immunosuppressed mouse hearts induced cardiac marker expression in retrovirus-infected cells within 2 weeks, although few cells showed striated muscle structures. To transduce GMT efficiently in vivo, we generated a polycistronic retrovirus expressing GMT separated by 2A "self-cleaving" peptides (3F2A). The 3F2A-induced cardiomyocyte-like cells in fibrotic tissue expressed sarcomeric α-actinin and cardiac troponin T and had clear cross striations. Quantitative RT-PCR also demonstrated that FACS-sorted 3F2A-transduced cells expressed cardiac-specific genes. CONCLUSIONS: GMT gene transfer induced cardiomyocyte-like cells in infarcted hearts.


Assuntos
Diferenciação Celular/genética , Fator de Transcrição GATA4/genética , Técnicas de Transferência de Genes , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Fatores de Regulação Miogênica/genética , Proteínas com Domínio T/genética , Animais , Diferenciação Celular/fisiologia , Fibroblastos/patologia , Fator de Transcrição GATA4/fisiologia , Proteínas de Fluorescência Verde/genética , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Camundongos Transgênicos , Modelos Animais , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Fatores de Regulação Miogênica/fisiologia , Regeneração/genética , Regeneração/fisiologia , Retroviridae/genética , Proteínas com Domínio T/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA