Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 15(2): e1007971, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30763305

RESUMEN

The Wilms' tumor suppressor gene (Wt1) encodes a zinc finger transcription factor that plays an essential role in the development of kidneys, gonads, spleen, adrenals and heart. Recent findings suggest that WT1 could also be playing physiological roles in adults. Systemic deletion of WT1 in mice provokes a severe deterioration of the exocrine pancreas, with mesothelial disruption, E-cadherin downregulation, disorganization of acinar architecture and accumulation of ascitic transudate. Despite this extensive damage, pancreatic stellate cells do not become activated and lose their canonical markers. We observed that pharmacological induction of pancreatitis in normal mice provokes de novo expression of WT1 in pancreatic stellate cells, concomitant with their activation. When pancreatitis was induced in mice after WT1 ablation, pancreatic stellate cells expressed WT1 and became activated, leading to a partial rescue of the acinar structure and the quiescent pancreatic stellate cell population after recovery from pancreatitis. We propose that WT1 modulates through the RALDH2/retinoic acid axis the restabilization of a part of the pancreatic stellate cell population and, indirectly, the repair of the pancreatic architecture, since quiescent pancreatic stellate cells are required for pancreas stability and repair. Thus, we suggest that WT1 plays novel and essential roles for the homeostasis of the adult pancreas and, through its upregulation in pancreatic stellate cells after a damage, for pancreatic regeneration. Due to the growing importance of the pancreatic stellate cells in physiological and pathophysiological conditions, these novel roles can be of translational relevance.


Asunto(s)
Genes del Tumor de Wilms , Páncreas/fisiología , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Aldehído Oxidorreductasas/metabolismo , Animales , Linaje de la Célula/genética , Ceruletida/toxicidad , Modelos Animales de Enfermedad , Epitelio/metabolismo , Expresión Génica , Homeostasis/genética , Homeostasis/fisiología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Estrelladas Pancreáticas/patología , Células Estrelladas Pancreáticas/fisiología , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/fisiopatología , Regeneración/genética , Regeneración/fisiología , Proteínas Represoras/deficiencia , Distribución Tisular , Investigación Biomédica Traslacional , Tretinoina/metabolismo , Proteínas WT1
2.
FASEB J ; 34(4): 5223-5239, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32068311

RESUMEN

The embryonic epicardium generates a population of epicardial-derived mesenchymal cells (EPDC) whose contribution to the coronary endothelium is minor or, according to some reports, negligible. We have compared four murine cell-tracing models related to the EPDC in order to elucidate this contribution. Cre recombinase was expressed under control of the promoters of the Wilms' tumor suppressor (Wt1), the cardiac troponin (cTnT), and the GATA5 genes, activating expression of the R26REYFP reporter. We have also used the G2 enhancer of the GATA4 gene as a driver due to its activation in the proepicardium. Recombination was found in most of the epicardium/EPDC in all cases. The contribution of these lineages to the cardiac endothelium was analyzed using confocal microscopy and flow cytometry. G2-GATA4 lineage cells are the most frequent in the endothelium, probably due to the recruitment of circulating endothelial progenitors. The contribution of the WT1 cell lineage increases along gestation due to further endothelial expression of WT1. GATA5 and cTnT lineages represent 4% of the cardiac endothelial cells throughout the gestation, probably standing for the actual EPDC contribution to the coronary endothelium. These results suggest caution when using a sole cell-tracing model to study the fate of the EPDC.


Asunto(s)
Linaje de la Célula , Vasos Coronarios/citología , Endotelio Vascular/citología , Pericardio/citología , Animales , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Integrasas , Ratones , Pericardio/embriología , Pericardio/metabolismo
3.
J Pathol ; 250(4): 362-373, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31875961

RESUMEN

Pancreatic heterotopia is defined as pancreatic tissue outside its normal location in the body and anatomically separated from the pancreas. In this work we have analyzed the stomach glandular epithelium of Gata4 flox/flox ; Pdx1-Cre mice (Gata4KO mice). We found that Gata4KO glandular epithelium displays an atypical morphology similar to the cornified squamous epithelium and exhibits upregulation of forestomach markers. The developing gastric units fail to form properly, and the glandular epithelial cells do not express markers of gastric gland in the absence of GATA4. Of interest, the developing glands of the Gata4KO stomach express pancreatic cell markers. Furthermore, a mass of pancreatic tissue located in the subserosa of the Gata4KO stomach is observed at adult stages. Heterotopic pancreas found in Gata4-deficient mice contains all three pancreatic cell lineages: ductal, acinar, and endocrine. Moreover, Gata4 expression is downregulated in ectopic pancreatic tissue of some human biopsy samples. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Células Epiteliales/patología , Factor de Transcripción GATA4/genética , Páncreas/patología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Mucosa Gástrica/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones Transgénicos , Organogénesis/fisiología
4.
IUBMB Life ; 72(1): 80-88, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31580534

RESUMEN

There is an urgent need for the development of novel therapeutics options for diabetic patients given the high prevalence of diabetes worldwide and that, currently, there is no cure for this disease. The transplantation of pancreatic islets that contain insulin-producing cells is a promising therapeutic alternative, particularly for type 1 diabetes. However, the shortage of organ donors constitutes a major limitation for this approach; thus, developing alternative sources of insulin-producing cells is of critical importance. In the last decade, our knowledge of the molecular mechanisms controlling embryonic pancreas development has significantly advanced. More importantly, this knowledge has provided the basis for the in vitro generation of insulin-producing cells from stem cells. Recent studies have revealed that GATA transcription factors are involved in various stages of pancreas formation and in the adult ß cell function. Here, we review the fundamental role of GATA transcription factors in pancreas morphogenesis and their association with congenital diseases associated with pancreas.


Asunto(s)
Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Páncreas/embriología , Enfermedades Pancreáticas/patología , Animales , Factores de Transcripción GATA/genética , Humanos , Páncreas/metabolismo , Enfermedades Pancreáticas/genética , Enfermedades Pancreáticas/metabolismo , Transducción de Señal
5.
Development ; 143(5): 774-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26811383

RESUMEN

Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/genética , Femenino , Eliminación de Gen , Corazón/embriología , Cardiopatías Congénitas/genética , Defectos del Tabique Interventricular/genética , Ventrículos Cardíacos , Humanos , Hibridación in Situ , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/fisiología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Morfogénesis/genética , Proteínas de Neoplasias/genética , Organogénesis , Análisis de Secuencia de ARN , Distribución Tisular , Transcripción Genética , Transposición de los Grandes Vasos/genética
6.
Proc Natl Acad Sci U S A ; 113(3): 656-61, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26739565

RESUMEN

Recent reports suggest that mammalian embryonic coronary endothelium (CoE) originates from the sinus venosus and ventricular endocardium. However, the contribution of extracardiac cells to CoE is thought to be minor and nonsignificant for coronary formation. Using classic (Wt1(Cre)) and previously undescribed (G2-Gata4(Cre)) transgenic mouse models for the study of coronary vascular development, we show that extracardiac septum transversum/proepicardium (ST/PE)-derived endothelial cells are required for the formation of ventricular coronary arterio-venous vascular connections. Our results indicate that at least 20% of embryonic coronary arterial and capillary endothelial cells derive from the ST/PE compartment. Moreover, we show that conditional deletion of the ST/PE lineage-specific Wilms' tumor suppressor gene (Wt1) in the ST/PE of G2-Gata4(Cre) mice and in the endothelium of Tie2(Cre) mice disrupts embryonic coronary transmural patterning, leading to embryonic death. Taken together, our results demonstrate that ST/PE-derived endothelial cells contribute significantly to and are required for proper coronary vascular morphogenesis.


Asunto(s)
Vasos Coronarios/embriología , Embrión de Mamíferos/citología , Células Endoteliales/citología , Tabiques Cardíacos/citología , Pericardio/citología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Vasos Coronarios/citología , Desarrollo Embrionario , Elementos de Facilitación Genéticos/genética , Transición Epitelial-Mesenquimal , Factor de Transcripción GATA4/metabolismo , Eliminación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/metabolismo , Ratones , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fenotipo , Proteínas WT1/metabolismo
7.
Dev Dyn ; 247(7): 924-933, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29708625

RESUMEN

The Wilms tumor suppressor gene (Wt1) encodes a transcription factor involved in the development of a number of organs, but the role played by Wt1 in pancreatic development is unknown. The pancreas contains a population of pancreatic stellate cells (PSC) very important for pancreatic physiology. We described elsewhere that hepatic stellate cells originate from the WT1-expressing liver mesothelium. Thus, we checked if the origin of PSCs was similar. WT1 expression is restricted to the pancreatic mesothelium. Between embryonic day (E) 10.5 and E15.5, this mesothelium gives rise to mesenchymal cells that contribute to a major part of the PSC and other cell types including endothelial cells. Most WT1 systemic mutants show abnormal localization of the dorsal pancreas within the mesentery and intestinal malrotation by E14.0. Embryos with conditional deletion of WT1 between E9.5 and E12.5 showed normal dorsal pancreatic bud and intestine, but the number of acini in the ventral bud was reduced approximately 30% by E16.5. Proliferation of acinar cells was reduced in WT1 systemic mutants, but pancreatic differentiation was not impaired. Thus, mesothelial-derived cells constitute an important subpopulation of pancreatic mesodermal cells. WT1 expression is not essential for pancreas development, although it influences intestinal rotation and correct localization of the dorsal pancreas within the mesogastrium. Developmental Dynamics 247:924-933, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Genes Supresores de Tumor/fisiología , Páncreas/crecimiento & desarrollo , Proteínas Represoras/genética , Células Acinares/citología , Animales , Proliferación Celular , Células Epiteliales , Epitelio , Intestinos/anatomía & histología , Mesodermo/citología , Ratones , Organogénesis , Células Estrelladas Pancreáticas , Proteínas Represoras/fisiología , Proteínas WT1
8.
Haematologica ; 102(4): 647-655, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28057738

RESUMEN

GATA transcription factors are expressed in the mesoderm and endoderm during development. GATA1-3, but not GATA4, are critically involved in hematopoiesis. An enhancer (G2) of the mouse Gata4 gene directs its expression throughout the lateral mesoderm and the allantois, beginning at embryonic day 7.5, becoming restricted to the septum transversum by embryonic day 10.5, and disappearing by midgestation. We have studied the developmental fate of the G2-Gata4 cell lineage using a G2-Gata4Cre;R26REYFP mouse line. We found a substantial number of YFP+ hematopoietic cells of lymphoid, myeloid and erythroid lineages in embryos. Fetal CD41+/cKit+/CD34+ and Lin-/cKit+/CD31+ YFP+ hematopoietic progenitors were much more abundant in the placenta than in the aorta-gonad-mesonephros area. They were clonogenic in the MethoCult assay and fully reconstituted hematopoiesis in myeloablated mice. YFP+ cells represented about 20% of the hematopoietic system of adult mice. Adult YFP+ hematopoietic stem cells constituted a long-term repopulating, transplantable population. Thus, a lineage of adult hematopoietic stem cells is characterized by the expression of GATA4 in their embryonic progenitors and probably by its extraembryonic (placental) origin, although GATA4 appeared not to be required for hematopoietic stem cell differentiation. Both lineages basically showed similar physiological behavior in normal mice, but clinically relevant properties of this particular hematopoietic stem cell population should be checked in physiopathological conditions.


Asunto(s)
Diferenciación Celular/genética , Factor de Transcripción GATA4/genética , Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mesodermo/citología , Placenta/citología , Animales , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Linaje de la Célula , Femenino , Factor de Transcripción GATA4/metabolismo , Inmunofenotipificación , Ratones , Ratones Transgénicos , Fenotipo , Embarazo
9.
Int J Mol Sci ; 18(6)2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28574454

RESUMEN

Consistent evidence from both experimental and human studies indicates that Type 2 diabetes mellitus (T2DM) is a complex disease resulting from the interaction of genetic, epigenetic, environmental, and lifestyle factors. Nutrients and dietary patterns are important environmental factors to consider in the prevention, development and treatment of this disease. Nutritional genomics focuses on the interaction between bioactive food components and the genome and includes studies of nutrigenetics, nutrigenomics and epigenetic modifications caused by nutrients. There is evidence supporting the existence of nutrient-gene and T2DM interactions coming from animal studies and family-based intervention studies. Moreover, many case-control, cohort, cross-sectional cohort studies and clinical trials have identified relationships between individual genetic load, diet and T2DM. Some of these studies were on a large scale. In addition, studies with animal models and human observational studies, in different countries over periods of time, support a causative relationship between adverse nutritional conditions during in utero development, persistent epigenetic changes and T2DM. This review provides comprehensive information on the current state of nutrient-gene interactions and their role in T2DM pathogenesis, the relationship between individual genetic load and diet, and the importance of epigenetic factors in influencing gene expression and defining the individual risk of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Dieta , Regulación de la Expresión Génica , Nutrigenómica , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dieta/efectos adversos , Epigénesis Genética , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Nutrigenómica/métodos
10.
Hepatology ; 59(6): 2358-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24415412

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo , Factor de Transcripción GATA4/fisiología , Cirrosis Hepática/metabolismo , Hígado/embriología , Animales , Intoxicación por Tetracloruro de Carbono/complicaciones , Línea Celular , Matriz Extracelular/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Integrasas , Cirrosis Hepática/etiología , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Fenotipo
11.
Cell Mol Life Sci ; 71(13): 2383-402, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24221136

RESUMEN

The field of pancreas development has markedly expanded over the last decade, significantly advancing our understanding of the molecular mechanisms that control pancreas organogenesis. This growth has been fueled, in part, by the need to generate new therapeutic approaches for the treatment of diabetes. The creation of sophisticated genetic tools in mice has been instrumental in this progress. Genetic manipulation involving activation or inactivation of genes within specific cell types has allowed the identification of many transcription factors (TFs) that play critical roles in the organogenesis of the pancreas. Interestingly, many of these TFs act at multiple stages of pancreatic development, and adult organ function or repair. Interaction with other TFs, extrinsic signals, and epigenetic regulation are among the mechanisms by which TFs may play context-dependent roles during pancreas organogenesis. Many of the pancreatic TFs directly regulate each other and their own expression. These combinatorial interactions generate very specific gene regulatory networks that can define the different cell lineages and types in the developing pancreas. Here, we review recent progress made in understanding the role of pancreatic TFs in mouse pancreas formation. We also summarize our current knowledge of human pancreas development and discuss developmental pancreatic TFs that have been associated with human pancreatic diseases.


Asunto(s)
Diabetes Mellitus/genética , Organogénesis/genética , Páncreas/crecimiento & desarrollo , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Diabetes Mellitus/terapia , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Mamíferos , Ratones
12.
Dev Biol ; 361(2): 439-49, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22056786

RESUMEN

The developing heart contains an inner tube of specialized endothelium known as endocardium, which performs multiple essential functions. In spite of the essential role of the endocardium in heart development and function, the transcriptional pathways that regulate its development remain largely undefined. GATA4 is a zinc finger transcription factor that is expressed in multiple cardiovascular lineages and is required for endocardial cushion development and embryonic viability, but the transcriptional pathways upstream of Gata4 in the endocardium and its derivatives in the endocardial cushions are unknown. Here, we describe a distal enhancer from the mouse Gata4 gene that is briefly active in multiple cardiac lineages early in cardiac development but restricts to the endocardium where it remains active through cardiogenesis. The activity of this Gata4 cardiac enhancer in transgenic embryos and in cultured aortic endothelial cells is dependent on four ETS sites. To identify which ETS transcription factors might be involved in Gata4 regulation via the ETS sites in the enhancer, we determined the expression profile of 24 distinct ETS factors in embryonic mouse hearts. Among multiple ETS transcripts present, ETS1, FLI1, ETV1, ETV5, ERG, and ETV6 were the most abundant in the early embryonic heart. We found that ETS1, FLI1, and ERG were strongly expressed in the heart at embryonic day 8.5 and that ETS1 and ERG bound to the endogenous Gata4 enhancer in cultured endothelial cells. Thus, these studies define the ETS expression profile in the early embryonic heart and identify an ETS-dependent enhancer from the Gata4 locus.


Asunto(s)
Elementos de Facilitación Genéticos , Factor de Transcripción GATA4/genética , Corazón/embriología , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Emparejamiento Base/genética , Secuencia de Bases , Sitios de Unión , Bovinos , Secuencia Conservada/genética , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos/genética , Ratones , Datos de Secuencia Molecular , Miocardio/citología , Miocardio/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción , Regulador Transcripcional ERG , Transgenes/genética
13.
Environ Pollut ; 316(Pt 2): 120633, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36370973

RESUMEN

Understanding the individual and global impact of pesticides on human physiology and the different stages of life is still a challenge in environmental health. We analyzed here whether administration of the organophosphate insecticide malathion before pregnancy could affect glucose homeostasis during pregnancy and, in addition, generate possible later consequences in mothers and offspring. For this, adult Wistar rats were allocated into two groups and were treated daily (intragastric) with malathion (14 or 140 mg/kg, body mass (bm)) for 21-25 days. Corn oil was used as vehicle in the Control group. Subgroups were defined based on the absence (nulliparous) or presence (pregnant) of a copulatory plug. Pregnant rats were followed by an additional period of 2 months after the term (post-term), without continuing malathion treatment. Fetuses and adult offspring of males and females were also evaluated. We ran an additional experimental design with rats exposed to malathion before pregnancy at a dose of 0.1 mg/kg bm. Malathion exposure resulted in glucose intolerance in the mothers during pregnancy and post-term period, regardless of the exposure dose. This was accompanied by increased visceral adipose tissue mass, dyslipidemia, unchanged pancreatic ß-cell mass, and varying insulin responses to glucose in vivo. The number of total newborns and birthweight was not affected by malathion exposure. Adult offspring from both sexes also became glucose-intolerant, regardless of the pesticide dose their dams were exposed to. This alteration could be associated with changes at the epigenomic level, as reduced hepatic mRNA content of DNA methylases and demethylases was found. We demonstrated that periconceptional exposure to malathion with doses aiming to mimic from work environment to indirect contamination predisposes progenitors and offspring rats to glucose intolerance. Thus, we conclude that subchronic exposure to malathion is a risk factor for gestational diabetes and prediabetes later in life.


Asunto(s)
Intolerancia a la Glucosa , Efectos Tardíos de la Exposición Prenatal , Recién Nacido , Embarazo , Masculino , Femenino , Ratas , Animales , Humanos , Malatión/toxicidad , Glucemia , Ratas Wistar , Homeostasis , Glucosa , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
14.
iScience ; 25(5): 104345, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35602948

RESUMEN

LRH-1/NR5A2 is implicated in islet morphogenesis postnatally, and its activation using the agonist BL001 protects islets against apoptosis, reverting hyperglycemia in mouse models of Type 1 Diabetes Mellitus. Islet transcriptome profiling revealed that the expression of PTGS2/COX2 is increased by BL001. Herein, we sought to define the role of LRH-1 in postnatal islet morphogenesis and chart the BL001 mode of action conferring beta cell protection. LRH-1 ablation within developing beta cells impeded beta cell proliferation, correlating with mouse growth retardation, weight loss, and hypoglycemia leading to lethality. LRH-1 deletion in adult beta cells abolished the BL001 antidiabetic action, correlating with beta cell destruction and blunted Ptgs2 induction. Islet PTGS2 inactivation led to reduced PGE2 levels and loss of BL001 protection against cytokines as evidenced by increased cytochrome c release and cleaved-PARP. The PTGER1 antagonist-ONO-8130-negated BL001-mediated islet survival. Our results define the LRH-1/PTGS2/PGE2/PTGER1 signaling axis as a key pathway mediating BL001 survival properties.

15.
Dev Biol ; 346(2): 346-55, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20692247

RESUMEN

The embryonic endoderm is a multipotent progenitor cell population that gives rise to the epithelia of the digestive and respiratory tracts, the liver and the pancreas. Among the transcription factors that have been shown to be important for endoderm development and gut morphogenesis is GATA4. Despite the important role of GATA4 in endoderm development, its transcriptional regulation is not well understood. In this study, we identified an intronic enhancer from the mouse Gata4 gene that directs expression to the definitive endoderm in the early embryo. The activity of this enhancer is initially broad in all endodermal progenitors, as demonstrated by fate mapping analysis using the Cre/loxP system, but becomes restricted to the dorsal foregut and midgut, and associated organs such as dorsal pancreas and stomach. The function of the intronic Gata4 enhancer is dependent upon a conserved Forkhead transcription factor-binding site, which is bound by recombinant FoxA2 in vitro. These studies identify Gata4 as a direct transcriptional target of FoxA2 in the hierarchy of the transcriptional regulatory network that controls the development of the definitive endoderm.


Asunto(s)
Endodermo/embriología , Elementos de Facilitación Genéticos/genética , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Intrones/genética , Animales , Secuencia de Bases , Sitios de Unión , Embrión de Mamíferos/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular
16.
Histochem Cell Biol ; 136(5): 595-607, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21932072

RESUMEN

The paired box transcription factor Pax8 is critical for development of the eye, thyroid gland as well as the urinary and reproductive organs. In adult, Pax8 overexpression is associated with kidney, ovarian and thyroid tumors and has emerged as a specific marker for these cancers. Recently, Pax8 expression was also reported in human pancreatic islets and in neuroendocrine tumors, identifying Pax8 as a novel member of the Pax family expressed in the pancreas. Herein, we sought to provide a comprehensive analysis of Pax8 expression during pancreogenesis and in adult islets. Immunohistochemical analysis using the most employed Pax8 polyclonal antibody revealed strong nuclear staining in the developing mouse pancreas and in mature human and mouse islets. Astonishingly, Pax8 mRNA in mouse islets was undetectable while human islets exhibited low levels. These discrepancies raised the possibility of antibody cross-reactivity. This premise was confirmed by demonstrating that the polyclonal Pax8 antibody also recognized the islet-enriched Pax6 protein both by Western blotting and immunohistochemistry. Thus, in islets polyclonal Pax8 staining corresponds mainly to Pax6. In order to circumvent this caveat, a novel Pax8 monoclonal antibody was used to re-evaluate whether Pax8 was indeed expressed in islets. Surprisingly, Pax8 was not detected in neither the developing pancreas or in mature islets. Reappraisal of pancreatic neuroendocrine tumors using this Pax8 monoclonal antibody exhibited no immunostaining as compared to the Pax8 polyclonal antibody. In conclusion, Pax8 is not expressed in the pancreas and cast doubts on the value of Pax8 as a pancreatic neuroendocrine tumor marker.


Asunto(s)
Islotes Pancreáticos/embriología , Tumores Neuroendocrinos/metabolismo , Factores de Transcripción Paired Box/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores de Tumor/metabolismo , Núcleo Celular/metabolismo , Reacciones Cruzadas , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Islotes Pancreáticos/metabolismo , Riñón/embriología , Riñón/metabolismo , Hígado/embriología , Hígado/metabolismo , Ratones , Tumores Neuroendocrinos/patología , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box/genética , Páncreas/embriología , Páncreas/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/metabolismo
17.
JCI Insight ; 6(23)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34699385

RESUMEN

In response to liver injury, hepatic stellate cells activate and acquire proliferative and contractile features. The regression of liver fibrosis appears to involve the clearance of activated hepatic stellate cells, either by apoptosis or by reversion toward a quiescent-like state, a process called deactivation. Thus, deactivation of active hepatic stellate cells has emerged as a novel and promising therapeutic approach for liver fibrosis. However, our knowledge of the master regulators involved in the deactivation and/or activation of fibrotic hepatic stellate cells is still limited. The transcription factor GATA4 has been previously shown to play an important role in embryonic hepatic stellate cell quiescence. In this work, we show that lack of GATA4 in adult mice caused hepatic stellate cell activation and, consequently, liver fibrosis. During regression of liver fibrosis, Gata4 was reexpressed in deactivated hepatic stellate cells. Overexpression of Gata4 in hepatic stellate cells promoted liver fibrosis regression in CCl4-treated mice. GATA4 induced changes in the expression of fibrogenic and antifibrogenic genes, promoting hepatic stellate cell deactivation. Finally, we show that GATA4 directly repressed EPAS1 transcription in hepatic stellate cells and that stabilization of the HIF2α protein in hepatic stellate cells leads to liver fibrosis.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/genética , Animales , Humanos , Cirrosis Hepática/patología , Ratones , Transfección
18.
Sci Rep ; 11(1): 8250, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859314

RESUMEN

Dietary fatty acids play a role in the pathogenesis of obesity-associated non-alcoholic fatty liver disease (NAFLD), which is associated with insulin resistance (IR). Fatty acid composition is critical for IR and subsequent NAFLD development. Extra-virgin olive oil (EVOO) is the main source of monounsaturated fatty acids (MUFA) in Mediterranean diets. This study examined whether EVOO-containing high fat diets may prevent diet-induced NAFLD using Ldlr-/-. Leiden mice. In female Ldlr-/-.Leiden mice, the effects of the following high fat diets (HFDs) were examined: a lard-based HFD (HFD-L); an EVOO-based HFD (HFD-EVOO); a phenolic compounds-rich EVOO HFD (HFD-OL). We studied changes in body weight (BW), lipid profile, transaminases, glucose homeostasis, liver pathology and transcriptome. Both EVOO diets reduced body weight (BW) and improved insulin sensitivity. The EVOOs did not improve transaminase values and increased LDL-cholesterol and liver collagen content. EVOOs and HFD-L groups had comparable liver steatosis. The profibrotic effects were substantiated by an up-regulation of gene transcripts related to glutathione metabolism, chemokine signaling and NF-kappa-B activation and down-regulation of genes relevant for fatty acid metabolism. Collectivelly, EVOO intake improved weight gain and insulin sensitivity but not liver inflammation and fibrosis, which was supported by changes in hepatic genes expression.


Asunto(s)
Peso Corporal/efectos de los fármacos , Resistencia a la Insulina , Obesidad/dietoterapia , Aceite de Oliva/farmacología , Receptores de LDL/genética , Animales , Dieta Alta en Grasa , Dieta Mediterránea , Femenino , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Noqueados , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo
19.
Mol Cell Biol ; 27(16): 5910-20, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17562853

RESUMEN

The MyoD family of basic helix-loop-helix (bHLH) transcription factors has the remarkable ability to induce myogenesis in vitro and in vivo. This myogenic specificity has been mapped to two amino acids in the basic domain, an alanine and threonine, referred to as the myogenic code. These essential determinants of myogenic specificity are conserved in all MyoD family members from worms to humans, yet their function in myogenesis is unclear. Induction of the muscle transcriptional program requires that MyoD be able to locate and stably bind to sequences present in the promoter regions of critical muscle genes. Recent studies have shown that MyoD binds to noncanonical E boxes in the myogenin gene, a critical locus required for myogenesis, through interactions with resident heterodimers of the HOX-TALE transcription factors Pbx1A and Meis1. In the present study, we show that the myogenic code is required for MyoD to bind to noncanonical E boxes in the myogenin promoter and for the formation of a tetrameric complex with Pbx/Meis. We also show that these essential determinants of myogenesis are sufficient to confer noncanonical E box binding to the E12 basic domain. Thus, these data show that noncanonical E box binding correlates with myogenic potential, and we speculate that the myogenic code residues in MyoD function as myogenic determinants via their role in noncanonical E box binding and recognition.


Asunto(s)
Elementos E-Box/genética , Desarrollo de Músculos/genética , Proteína MioD/metabolismo , Animales , Secuencia de Bases , ADN/metabolismo , Dimerización , Proteínas de Homeodominio/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteína MioD/genética , Miogenina/genética , Proteínas de Neoplasias/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Transcripción Genética , Activación Transcripcional/genética
20.
Adv Exp Med Biol ; 654: 59-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217494

RESUMEN

Over the last years, there has been great success in driving stem cells toward insulin-expressing cells. However, the protocols developed to date have some limitations, such as low reliability and low insulin production. The most successful protocols used for generation of insulin-producing cells from stem cells mimic in vitro pancreatic organogenesis by directing the stem cells through stages that resemble several pancreatic developmental stages. Islet cell fate is coordinated by a complex network of inductive signals and regulatory transcription factors that, in a combinatorial way, determine pancreatic organ specification, differentiation, growth, and lineage. Together, these signals and factors direct the progression from multipotent progenitor cells to mature pancreatic cells. Later in development and adult life, several of these factors also contribute to maintain the differentiated phenotype of islet cells. A detailed understanding of the processes that operate in the pancreas during embryogenesis will help us to develop a suitable source of cells for diabetes therapy. In this chapter, we will discuss the main transcription factors involved in pancreas specification and beta-cell formation.


Asunto(s)
Biología Evolutiva/métodos , Islotes Pancreáticos/citología , Islotes Pancreáticos/patología , Páncreas/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula , Sistema Endocrino/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Ratones , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Páncreas/embriología , Células Madre/citología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA