Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circulation ; 138(10): 1025-1038, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-29567669

RESUMEN

BACKGROUND: Bicuspid aortic valve (BAV), the most common congenital heart defect affecting 1% to 2% of the population, is a major risk factor for premature aortic valve disease and accounts for the majority of valve replacement. The genetic basis and mechanisms of BAV etiology and pathogenesis remain largely undefined. METHODS: Cardiac structure and function was assessed in mice lacking a Gata6 allele. Human GATA6 gene variants were analyzed in 452 BAV cases from the BAV consortium and 1849 controls from the Framingham GWAS (Genome Wide Association Study). GATA6 expression was determined in mice and human tissues using quantitative real-time polymerase chain reaction and immunohistochemistry. Mechanistic studies were carried out in cultured cells. RESULTS: Gata6 heterozygous mice have highly penetrant right-left (RL)-type BAV, the most frequent type in humans. GATA6 transcript levels are lower in human BAV compared with normal tricuspid valves. Mechanistically, Gata6 haploinsufficiency disrupts valve remodeling and extracellular matrix composition through dysregulation of important signaling molecules, including matrix metalloproteinase 9. Cell-specific inactivation of Gata6 reveals an essential role for GATA6 in secondary heart field myocytes because loss of 1 Gata6 allele from Isl- 1-positive cells-but not from endothelial or neural crest cells-recapitulates the phenotype of Gata6 heterozygous mice. CONCLUSIONS: The data identify a new cellular and molecular mechanism underlying BAV. The availability of an animal model for the most frequent human BAV opens the way for the elucidation of BAV pathogenesis and the development of much needed therapies.


Asunto(s)
Válvula Aórtica/anomalías , Válvula Aórtica/metabolismo , Factor de Transcripción GATA6/genética , Haploinsuficiencia , Enfermedades de las Válvulas Cardíacas/genética , Animales , Válvula Aórtica/patología , Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Factor de Transcripción GATA6/deficiencia , Predisposición Genética a la Enfermedad , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Enfermedades de las Válvulas Cardíacas/fisiopatología , Heterocigoto , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
2.
Reproduction ; 154(4): 455-467, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28710293

RESUMEN

Testicular Leydig cells produce androgens essential for proper male reproductive development and fertility. Here, we describe a new Leydig cell ablation model based on Cre/Lox recombination of mouse Gata4 and Gata6, two genes implicated in the transcriptional regulation of steroidogenesis. The testicular interstitium of adult Gata4flox/flox ; Gata6flox/flox mice was injected with adenoviral vectors encoding Cre + GFP (Ad-Cre-IRES-GFP) or GFP alone (Ad-GFP). The vectors efficiently and selectively transduced Leydig cells, as evidenced by GFP reporter expression. Three days after Ad-Cre-IRES-GFP injection, expression of androgen biosynthetic genes (Hsd3b1, Cyp17a1 and Hsd17b3) was reduced, whereas expression of another Leydig cell marker, Insl3, was unchanged. Six days after Ad-Cre-IRES-GFP treatment, the testicular interstitium was devoid of Leydig cells, and there was a concomitant loss of all Leydig cell markers. Chromatin condensation, nuclear fragmentation, mitochondrial swelling, and other ultrastructural changes were evident in the degenerating Leydig cells. Liquid chromatography-tandem mass spectrometry demonstrated reduced levels of androstenedione and testosterone in testes from mice injected with Ad-Cre-IRES-GFP. Late effects of treatment included testicular atrophy, infertility and the accumulation of lymphoid cells in the testicular interstitium. We conclude that adenoviral-mediated gene delivery is an expeditious way to probe Leydig cell function in vivo Our findings reinforce the notion that GATA factors are key regulators of steroidogenesis and testicular somatic cell survival.Free Finnish abstract: A Finnish translation of this abstract is freely available at http://www.reproduction-online.org/content/154/4/455/suppl/DC2.


Asunto(s)
Adenoviridae/genética , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Vectores Genéticos , Células Intersticiales del Testículo/metabolismo , Transducción Genética , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Supervivencia Celular , Femenino , Fertilidad , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Genotipo , Hormonas Esteroides Gonadales/biosíntesis , Insulina/genética , Insulina/metabolismo , Integrasas/genética , Células Intersticiales del Testículo/ultraestructura , Masculino , Ratones Noqueados , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Fenotipo , Embarazo , Progesterona Reductasa/genética , Progesterona Reductasa/metabolismo , Proteínas/genética , Proteínas/metabolismo , Transducción de Señal , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Esteroide Isomerasas/genética , Esteroide Isomerasas/metabolismo , Factores de Tiempo
3.
Nat Cell Biol ; 19(6): 603-613, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28504705

RESUMEN

The epidermis is maintained by multiple stem cell populations whose progeny differentiate along diverse, and spatially distinct, lineages. Here we show that the transcription factor Gata6 controls the identity of the previously uncharacterized sebaceous duct (SD) lineage and identify the Gata6 downstream transcription factor network that specifies a lineage switch between sebocytes and SD cells. During wound healing differentiated Gata6+ cells migrate from the SD into the interfollicular epidermis and dedifferentiate, acquiring the ability to undergo long-term self-renewal and differentiate into a much wider range of epidermal lineages than in undamaged tissue. Our data not only demonstrate that the structural and functional complexity of the junctional zone is regulated by Gata6, but also reveal that dedifferentiation is a previously unrecognized property of post-mitotic, terminally differentiated cells that have lost contact with the basement membrane. This resolves the long-standing debate about the contribution of terminally differentiated cells to epidermal wound repair.


Asunto(s)
Desdiferenciación Celular , Epidermis/metabolismo , Factor de Transcripción GATA6/metabolismo , Glándulas Sebáceas/metabolismo , Células Madre/metabolismo , Cicatrización de Heridas , Heridas y Lesiones/metabolismo , Animales , Linaje de la Célula , Movimiento Celular , Plasticidad de la Célula , Autorrenovación de las Células , Células Cultivadas , Modelos Animales de Enfermedad , Epidermis/patología , Femenino , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Glándulas Sebáceas/patología , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Heridas y Lesiones/genética , Heridas y Lesiones/patología
4.
Gastroenterology ; 147(5): 1119-33.e4, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25127677

RESUMEN

BACKGROUND & AIMS: Although smoking is a leading risk factor for pancreatic ductal adenocarcinoma (PDAC), little is known about the mechanisms by which smoking promotes initiation or progression of PDAC. METHODS: We studied the effects of nicotine administration on pancreatic cancer development in Kras(+/LSLG12Vgeo);Elas-tTA/tetO-Cre (Ela-KRAS) mice, Kras(+/LSLG12D);Trp53+/LSLR172H;Pdx-1-Cre (KPC) mice (which express constitutively active forms of KRAS), and C57/B6 mice. Mice were given nicotine for up to 86 weeks to produce blood levels comparable with those of intermediate smokers. Pancreatic tissues were collected and analyzed by immunohistochemistry and reverse transcriptase polymerase chain reaction; cells were isolated and assayed for colony and sphere formation and gene expression. The effects of nicotine were also evaluated in primary pancreatic acinar cells isolated from wild-type, nAChR7a(-/-), Trp53(-/-), and Gata6(-/-);Trp53(-/-) mice. We also analyzed primary PDAC cells that overexpressed GATA6 from lentiviral expression vectors. RESULTS: Administration of nicotine accelerated transformation of pancreatic cells and tumor formation in Ela-KRAS and KPC mice. Nicotine induced dedifferentiation of acinar cells by activating AKT-ERK-MYC signaling; this led to inhibition of Gata6 promoter activity, loss of GATA6 protein, and subsequent loss of acinar differentiation and hyperactivation of oncogenic KRAS. Nicotine also promoted aggressiveness of established tumors as well as the epithelial-mesenchymal transition, increasing numbers of circulating cancer cells and their dissemination to the liver, compared with mice not exposed to nicotine. Nicotine induced pancreatic cells to acquire gene expression patterns and functional characteristics of cancer stem cells. These effects were markedly attenuated in K-Ras(+/LSL-G12D);Trp53(+/LSLR172H);Pdx-1-Cre mice given metformin. Metformin prevented nicotine-induced pancreatic carcinogenesis and tumor growth by up-regulating GATA6 and promoting differentiation toward an acinar cell program. CONCLUSIONS: In mice, nicotine promotes pancreatic carcinogenesis and tumor development via down-regulation of Gata6 to induce acinar cell dedifferentiation.


Asunto(s)
Células Acinares/efectos de los fármacos , Carcinoma Ductal Pancreático/inducido químicamente , Desdiferenciación Celular/efectos de los fármacos , Factor de Transcripción GATA6/metabolismo , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Páncreas/efectos de los fármacos , Neoplasias Pancreáticas/inducido químicamente , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Acinares/metabolismo , Células Acinares/patología , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/prevención & control , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Metformina/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutación , Células Neoplásicas Circulantes/efectos de los fármacos , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética
5.
J Exp Med ; 211(8): 1525-31, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-25024137

RESUMEN

The transcription factor Gata6 regulates proliferation and differentiation of epithelial and endocrine cells and cancers. Among hematopoietic cells, Gata6 is expressed selectively in resident peritoneal macrophages. We thus examined whether the loss of Gata6 in the macrophage compartment affected peritoneal macrophages, using Lyz2-Cre x Gata6(flox/flox) mice to tackle this issue. In Lyz2-Cre x Gata6(flox/flox) mice, the resident peritoneal macrophage compartment, but not macrophages in other organs, was contracted, with only a third the normal number of macrophages remaining. Heightened rates of death explained the marked decrease in peritoneal macrophage observed. The metabolism of the remaining macrophages was skewed to favor oxidative phosphorylation and alternative activation markers were spontaneously and selectively induced in Gata6-deficient macrophages. Gene expression profiling revealed perturbed metabolic regulators, including aspartoacylase (Aspa), which facilitates generation of acetyl CoA. Mutant mice lacking functional Aspa phenocopied the higher propensity to death and led to a contraction of resident peritoneal macrophages. Thus, Gata6 regulates differentiation, metabolism, and survival of resident peritoneal macrophages.


Asunto(s)
Amidohidrolasas/metabolismo , Factor de Transcripción GATA6/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/enzimología , Animales , Apoptosis/genética , Recuento de Células , Supervivencia Celular , Factor de Transcripción GATA6/deficiencia , Regulación de la Expresión Génica , Activación de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular
6.
Am J Pathol ; 182(6): 2391-406, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583651

RESUMEN

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease characterized by pulmonary vasculopathy with elevation of pulmonary artery pressure, often culminating in right ventricular failure. GATA-6, a member of the GATA family of zinc-finger transcription factors, is highly expressed in quiescent vasculature and is frequently lost during vascular injury. We hypothesized that endothelial GATA-6 may play a critical role in the molecular mechanisms underlying endothelial cell (EC) dysfunction in PAH. Here we report that GATA-6 is markedly reduced in pulmonary ECs lining both occluded and nonoccluded vessels in patients with idiopathic and systemic sclerosis-associated PAH. GATA-6 transcripts are also rapidly decreased in rodent PAH models. Endothelial GATA-6 is a direct transcriptional regulator of genes controlling vascular tone [endothelin-1, endothelin-1 receptor type A, and endothelial nitric oxide synthase (eNOS)], pro-inflammatory genes, CX3CL1 (fractalkine), 5-lipoxygenease-activating protein, and markers of vascular remodeling, including PAI-1 and RhoB. Mice with the genetic deletion of GATA-6 in ECs (Gata6-KO) spontaneously develop elevated pulmonary artery pressure and increased vessel muscularization, and these features are further exacerbated in response to hypoxia. Furthermore, innate immune cells including macrophages (CD11b(+)/F4/80(+)), granulocytes (Ly6G(+)/CD45(+)), and dendritic cells (CD11b(+)/CD11c(+)) are significantly increased in normoxic Gata6-KO mice. Together, our findings suggest a critical role of endothelial GATA-6 deficiency in development and disease progression in PAH.


Asunto(s)
Endotelio Vascular/metabolismo , Factor de Transcripción GATA6/deficiencia , Hipertensión Pulmonar/metabolismo , Adaptación Fisiológica/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Casos y Controles , Enfermedad Crónica , Progresión de la Enfermedad , Regulación hacia Abajo/fisiología , Células Endoteliales/fisiología , Hipertensión Pulmonar Primaria Familiar , Factor de Transcripción GATA6/metabolismo , Factor de Transcripción GATA6/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Pulmón/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Neumonía/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Esclerodermia Sistémica/complicaciones
7.
Gut ; 62(10): 1481-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23002247

RESUMEN

OBJECTIVES: Previous studies have suggested an important role of the transcription factor Gata6 in endocrine pancreas, while GATA6 haploinsufficient inactivating mutations cause pancreatic agenesis in humans. We aimed to analyse the effects of Gata6 inactivation on pancreas development and function. DESIGN: We deleted Gata6 in all epithelial cells in the murine pancreas at the onset of its development. Acinar proliferation, apoptosis, differentiation and exocrine functions were assessed using reverse transcriptase quantitative PCR (RT-qPCR), chromatin immunoprecipitation, immunohistochemistry and enzyme assays. Adipocyte transdifferentiation was assessed using electron microscopy and genetic lineage tracing. RESULTS: Gata6 is expressed in all epithelial cells in the adult mouse pancreas but it is only essential for exocrine pancreas homeostasis: while dispensable for pancreatic development after e10.5, it is required for complete acinar differentiation, for establishment of polarity and for the maintenance of acinar cells in the adult. Gata6 regulates directly the promoter of genes coding for digestive enzymes and the transcription factors Rbpjl and Mist1. Upon pancreas-selective Gata6 inactivation, massive loss of acinar cells and fat replacement take place. This is accompanied by increased acinar apoptosis and proliferation, acinar-to-ductal metaplasia and adipocyte transdifferentiation. By contrast, the endocrine pancreas is spared. CONCLUSIONS: Our data show that Gata6 is required for the complete differentiation of acinar cells through multiple transcriptional regulatory mechanisms. In addition, it is required for the maintenance of the adult acinar cell compartment. Our studies suggest that GATA6 alterations may contribute to diseases of the human adult exocrine pancreas.


Asunto(s)
Células Acinares/citología , Factor de Transcripción GATA6/fisiología , Páncreas Exocrino/citología , Células Acinares/patología , Células Acinares/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Femenino , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Eliminación de Gen , Masculino , Metaplasia/genética , Metaplasia/patología , Ratones , Ratones Noqueados , Páncreas Exocrino/crecimiento & desarrollo , Páncreas Exocrino/patología , Páncreas Exocrino/fisiología
8.
J Clin Invest ; 122(10): 3516-28, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23006325

RESUMEN

Pancreatic agenesis is a human disorder caused by defects in pancreas development. To date, only a few genes have been linked to pancreatic agenesis in humans, with mutations in pancreatic and duodenal homeobox 1 (PDX1) and pancreas-specific transcription factor 1a (PTF1A) reported in only 5 families with described cases. Recently, mutations in GATA6 have been identified in a large percentage of human cases, and a GATA4 mutant allele has been implicated in a single case. In the mouse, Gata4 and Gata6 are expressed in several endoderm-derived tissues, including the pancreas. To analyze the functions of GATA4 and/or GATA6 during mouse pancreatic development, we generated pancreas-specific deletions of Gata4 and Gata6. Surprisingly, loss of either Gata4 or Gata6 in the pancreas resulted in only mild pancreatic defects, which resolved postnatally. However, simultaneous deletion of both Gata4 and Gata6 in the pancreas caused severe pancreatic agenesis due to disruption of pancreatic progenitor cell proliferation, defects in branching morphogenesis, and a subsequent failure to induce the differentiation of progenitor cells expressing carboxypeptidase A1 (CPA1) and neurogenin 3 (NEUROG3). These studies address the conserved and nonconserved mechanisms underlying GATA4 and GATA6 function during pancreas development and provide a new mouse model to characterize the underlying developmental defects associated with pancreatic agenesis.


Asunto(s)
Factor de Transcripción GATA4/fisiología , Factor de Transcripción GATA6/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Organogénesis/genética , Páncreas/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Sitios de Unión , Carboxipeptidasas A/análisis , Diferenciación Celular , División Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Endodermo/metabolismo , Células Epiteliales/patología , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Técnicas de Silenciamiento del Gen , Genotipo , Edad Gestacional , Hiperglucemia/congénito , Hiperglucemia/genética , Insulina/metabolismo , Secreción de Insulina , Ratones , Proteínas del Tejido Nervioso/análisis , Especificidad de Órganos , Páncreas/anomalías , Páncreas/patología , Transcripción Genética
9.
J Clin Invest ; 122(10): 3504-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23006330

RESUMEN

Recently, heterozygous mutations in GATA6 have been found in neonatal diabetic patients with failed pancreatic organogenesis. To investigate the roles of GATA4 and GATA6 in mouse pancreas organogenesis, we conditionally inactivated these genes within the pancreas. Single inactivation of either gene did not have a major impact on pancreas formation, indicating functional redundancy. However, double Gata4/Gata6 mutant mice failed to develop pancreata, died shortly after birth, and displayed hyperglycemia. Morphological defects in Gata4/Gata6 mutant pancreata were apparent during embryonic development, and the epithelium failed to expand as a result of defects in cell proliferation and differentiation. The number of multipotent pancreatic progenitors, including PDX1+ cells, was reduced in the Gata4/Gata6 mutant pancreatic epithelium. Remarkably, deletion of only 1 Gata6 allele on a Gata4 conditional knockout background severely reduced pancreatic mass. In contrast, a single WT allele of Gata4 in Gata6 conditional knockout mice was sufficient for normal pancreatic development, indicating differential contributions of GATA factors to pancreas formation. Our results place GATA factors at the top of the transcriptional network hierarchy controlling pancreas organogenesis.


Asunto(s)
Factor de Transcripción GATA4/fisiología , Factor de Transcripción GATA6/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Organogénesis/genética , Páncreas/embriología , Transactivadores/genética , Animales , Sitios de Unión , Diferenciación Celular , División Celular , Células Epiteliales/patología , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Genotipo , Proteínas de Homeodominio/biosíntesis , Hiperglucemia/congénito , Hiperglucemia/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Páncreas/anomalías , Páncreas/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Transactivadores/biosíntesis , Transcripción Genética
10.
Dev Biol ; 358(2): 368-78, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21839733

RESUMEN

Members of the GATA family of transcription factors are critical regulators of heart development and mutations in 2 of them, GATA4 and GATA6 are associated with outflow tract and septal defects in human. The heart expresses 3 GATA factors, GATA4, 5 and 6 in a partially overlapping pattern. Here, we report that compound Gata4/Gata5 and Gata5/Gata6 mutants die embryonically or perinatally due to severe congenital heart defects. Almost all Gata4(+/-)Gata5(+/-) mutant embryos have double outlet right ventricles (DORV), large ventricular septal defects (VSD) as well as hypertrophied mitral and tricuspid valves. Only 25% of double compound Gata4/Gata5 heterozygotes survive to adulthood and these mice have aortic stenosis. Compound loss of a Gata5 and a Gata6 allele also leads to DORVs associated with subaortic VSDs. Expression of several transcription factors important for endocardial and myocardial cell differentiation, such as Tbx20, Mef2c, Hey1 and Hand2, was reduced in compound heterozygote embryos. These findings suggest the existence of important genetic interactions between Gata5 and the 2 other cardiac GATA factors in endocardial cushion formation and outflow tract morphogenesis. The data identify GATA5 as a potential genetic modifier of congenital heart disease and provide insight for elucidating the genetic basis of an important class of human birth defects.


Asunto(s)
Corazón Fetal/embriología , Corazón Fetal/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA5/metabolismo , Factor de Transcripción GATA6/metabolismo , Animales , Estenosis de la Válvula Aórtica/embriología , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Femenino , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA5/deficiencia , Factor de Transcripción GATA5/genética , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/metabolismo , Heterocigoto , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Dev Dyn ; 237(10): 2820-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18816845

RESUMEN

The derivation of the primitive endoderm layer from the pluripotent cells of the inner cell mass is one of the earliest differentiation and morphogenic events in embryonic development. GATA4 and GATA6 are the key transcription factors in the formation of extraembryonic endoderms, but their specific contribution to the derivation of each endoderm lineage needs clarification. We further analyzed the dynamic expression and mutant phenotypes of GATA6 in early mouse embryos. GATA6 and GATA4 are both expressed in primitive endoderm cells initially. At embryonic day (E) 5.0, parietal endoderm cells continue to express both GATA4 and GATA6; however, visceral endoderm cells express GATA4 but exhibit a reduced expression of GATA6. By and after E5.5, visceral endoderm cells no longer express GATA6. We also found that GATA6 null embryos did not form a morphologically recognizable primitive endoderm layer, and subsequently failed to form visceral and parietal endoderms. Thus, the current study establishes that GATA6 is essential for the formation of primitive endoderm, at a much earlier stage then previously recognized, and expression of GATA6 discriminates parietal endoderm from visceral endoderm lineages.


Asunto(s)
Endodermo/embriología , Endodermo/metabolismo , Factor de Transcripción GATA6/metabolismo , Animales , Blastocisto/metabolismo , Diferenciación Celular , Línea Celular , Transferencia de Embrión , Células Madre Embrionarias , Femenino , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Factores de Tiempo
13.
Dev Biol ; 317(2): 614-9, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18400219

RESUMEN

Despite significant advances in identifying signaling molecules that induce cardiogenesis in mammals, the transcription factors that control the onset of cardiac myocyte gene expression have remained elusive. Candidates include the zinc finger transcription factors GATA binding proteins 4 and 6 (GATA4, GATA6). The individual loss of either protein in mice results in lethality prior to the onset of heart development due to defects in the extra-embryonic endoderm; however, when this extra-embryonic deficiency is circumvented using tetraploid embryo complementation, cardiac myocyte differentiation initiates normally. Here we show that these factors have redundant roles in controlling the onset of cardiac myocyte differentiation. As a consequence, Gata4(-/-)Gata6(-/-) embryos completely lack hearts, although second heart field progenitor cells are still generated. Our data support a model whereby GATA4 or GATA6 are essential for expression of the network of transcription factors that regulate the onset of cardiac myocyte gene expression during mammalian development.


Asunto(s)
Diferenciación Celular/fisiología , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/embriología , Miocitos Cardíacos/citología , Animales , Endodermo/fisiología , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA6/deficiencia , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...