Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Biol Toxicol ; 39(4): 1657-1676, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36029422

RESUMEN

Key regulatory decisions during cleavage divisions in mammalian embryogenesis determine the fate of preimplantation embryonic cells. Single-cell RNA sequencing of early-stage-2-cell, 4-cell, and 8-cell-blastomeres show that the aryl hydrocarbon receptor (AHR), traditionally considered as an environmental sensor, directs blastomere differentiation. Disruption of AHR functions in Ahr knockout embryos or in embryos from dams exposed to dioxin, the prototypic xenobiotic AHR agonist, significantly impairs blastocyst formation, causing repression and loss of transcriptional heterogeneity of OCT4 and CDX2 and incidence of nonspecific downregulation of pluripotency. Trajectory-the path of differentiation-and gene variability analyses further confirm that deregulation of OCT4 functions and changes of transcriptional heterogeneity resulting from disruption of AHR functions restrict the emergence of differentiating blastomeres in 4-cell embryos. It appears that AHR directs the differentiation of progenitor blastomeres and that disruption of preimplantation AHR functions may significantly perturb embryogenesis leading to long-lasting conditions at the heart of disease in offspring's adulthood.


Asunto(s)
Blastómeros , Receptores de Hidrocarburo de Aril , Animales , Ratones , Diferenciación Celular , Desarrollo Embrionario , Mamíferos , Receptores de Hidrocarburo de Aril/genética
2.
Toxicol Sci ; 182(1): 1-9, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34009372

RESUMEN

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor well-known for its adaptive role as a sensor of environmental toxicants and mediator of the metabolic detoxification of xenobiotic ligands. In addition, a growing body of experimental data has provided indisputable evidence that the AHR regulates critical functions of cell physiology and embryonic development. Recent studies have shown that the naïve AHR-that is, unliganded to xenobiotics but activated endogenously-has a crucial role in maintenance of embryonic stem cell pluripotency, tissue repair, and regulation of cancer stem cell stemness. Depending on the cellular context, AHR silences the expression of pluripotency genes Oct4 and Nanog and potentiates differentiation, whereas curtailing cellular plasticity and stemness. In these processes, AHR-mediated contextual responses and outcomes are dictated by changes of interacting partners in signaling pathways, gene networks, and cell-type-specific genomic structures. In this review, we focus on AHR-mediated changes of genomic architecture as an emerging mechanism for the AHR to regulate gene expression at the transcriptional level. Collective evidence places this receptor as a physiological hub connecting multiple biological processes whose disruption impacts on embryonic development, tissue repair, and maintenance or loss of stemness.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Hidrocarburo de Aril , Diferenciación Celular , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , Humanos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
3.
Toxicol Sci ; 178(2): 325-337, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33017471

RESUMEN

Congenital heart disease (CHD), the leading birth defect worldwide, has a largely unknown etiology, likely to result from complex interactions between genetic and environmental factors during heart development, at a time when the heart adapts to diverse physiological and pathophysiological conditions. Crucial among these is the regulation of cardiomyocyte development and postnatal maturation, governed by dynamic changes in DNA methylation. Previous work from our laboratory has shown that exposure to the environmental toxicant tetrachlorodibenzo-p-dioxin (TCDD) disrupts several molecular networks responsible for heart development and function. To test the hypothesis that the disruption caused by TCDD in the heart results from changes in DNA methylation and gene expression patterns of cardiomyocytes, we established a stable mouse embryonic stem cell line expressing a puromycin resistance selectable marker under control of the cardiomyocyte-specific Nkx2-5 promoter. Differentiation of these cells in the presence of puromycin induces the expression of a large suite of cardiomyocyte-specific markers. To assess the consequences of TCDD treatment on gene expression and DNA methylation in these cardiomyocytes, we subjected them to transcriptome and methylome analyses in the presence of TCDD. Unlike control cardiomyocytes maintained in vehicle, the TCDD-treated cardiomyocytes showed extensive gene expression changes, with a significant correlation between differential RNA expression and DNA methylation in 111 genes, many of which are key elements of pathways that regulate cardiovascular development and function. Our findings provide an important clue toward the elucidation of the complex interactions between genetic and epigenetic mechanisms after developmental TCDD exposure that may contribute to CHD.


Asunto(s)
Metilación de ADN , Dioxinas/toxicidad , Células Madre Embrionarias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Dibenzodioxinas Policloradas , Animales , Línea Celular , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Dibenzodioxinas Policloradas/toxicidad
4.
Curr Opin Toxicol ; 2: 1-7, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28920102

RESUMEN

Recent evidence from embryonic stem cells suggests that the aryl hydrocarbon receptor (AHR) plays a central role in the regulation of pluripotency, a short-lived property of cells in the early blastula inner cell mass (ICM). Four key observations support this conclusion. The first is the temporal association between upregulation of AHR expression and the onset of cell differentiation, which argues for the AHR as a determinant of cell fate decisions. The second is the repression of the pluripotency factors OCT4 and NANOG by the AHR, which depresses their function and contributes to the cell's exit from pluripotency. The third is the temporal association between changes in global DNA methylation and stage-dependent AHR expression, which parallel each other during embryonic development, suggesting that AHR helps configure a repressive chromatin structure that controls differentiation. The fourth is the incidence of early developmental aberrations that take place in Ahr-null mice and cause the disruption of their embryonic program, which is likely to be a consequence of the loss of pluripotency of the Ahr-/- ICM cells. In this short review, we will focus on the modulation of pluripotency as a novel function of the AHR, and on the potentially detrimental developmental outcomes that may result from exposure to environmental toxicants. This line of enquiry brings us to the tantalizing conclusion that by activating mechanisms that modulate pluripotency, AHR regulates embryonic development. The likelihood that exposure to environmental AHR ligands might disrupt developmental processes is a reasonable corollary to this conclusion.

5.
Sci Rep ; 7(1): 6306, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28740224

RESUMEN

Pain is a distressing symptom of Parkinson disease (PD). We aim to determine whether the genetic variants of chronic pain-related genes contribute to pain in PD patients. We included 418 PD patients and evaluated pain severity on King's PD pain scale. We genotyped rs6267, rs6269, rs4633, rs4818 and rs4680 of COMT, rs6746030 of SCN9A, and rs1799971 of OPRM1. In total, 193 participants (46.2%) experienced pain. Compared to pain-free PD patients, PD patients with pain had an earlier age of onset, longer disease duration, and higher depression and motor severity (P < 0.01). The frequencies of COMT rs4680 "A" allele were higher in PD patients with pain than those without pain (46.1% vs. 31.1%, P < 0.01). Pain severity was significantly associated with disease duration (P = 0.02), and COMT rs6267 T allele (P < 0.01). We stratified PD by status of depression and the association between COMT rs6267 "GT" genotype and pain severity remained significant (P < 0.01). Furthermore, pain severity was significantly higher in participants having COMT rs4680 "GG" and "GA" genpotypes than those having "AA" genotype (P = 0.04). We concluded that depression and COMT rs4680 "GG" and "GA" genotypes and COMT rs6267 "GT" genotype contribute to pain in PD patients.


Asunto(s)
Catecol O-Metiltransferasa/genética , Dolor Crónico/genética , Depresión/complicaciones , Enfermedad de Parkinson/complicaciones , Polimorfismo de Nucleótido Simple , Edad de Inicio , Anciano , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.7/genética , Dimensión del Dolor , Receptores Opioides mu/genética , Índice de Severidad de la Enfermedad
6.
Stem Cells ; 34(12): 2825-2839, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27374890

RESUMEN

Lack of cell cycle checkpoints and uninterrupted passage through S-phase continuously renew the embryonic stem (ES) cell population and maintain pluripotency. Here, we show that to regulate mitotic progression and pluripotency ES cells must keep the aryl hydrocarbon receptor (AHR), an environmental sensor and transcriptional regulator, in a persistent state of repression. This repression, however, is not always absolute, causing the AHR to fluctuate between reversible states of expression and repression, with a fraction of the cells escaping repression at any one time. Cells that escape AHR repression exhibit reduced levels of the pluripotency factors OCT4 and SOX2 and show an extended mitotic traverse time due to AHR-dependent MID1 repression and the subsequent disruption of the MID1-PP2A-CDC25B-CDK1 signaling pathway that regulates mitosis. Unlike the bulk of the cell population that differentiates into cardiomyocytes upon stimulation, AHR-expressing ES cells restrict cardiogenesis and commit to a neuroglia cell fate. It appears that the untimely expression of the Ahr gene needs to be repressed to maintain ES cell mitotic progression and prevent premature loss of pluripotency. Stem Cells 2016;34:2825-2839.


Asunto(s)
Mitosis , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Regulación de la Expresión Génica , Ratones , Mitosis/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Receptores de Hidrocarburo de Aril/genética , Proteínas Represoras/genética , Fase S/genética , Transducción de Señal/genética
7.
Toxicol Sci ; 149(2): 346-57, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26572662

RESUMEN

The AHR is a ligand-activated transcription factor that mediates gene-environment interactions. Genome-wide expression profiling during differentiation of mouse ES cells into cardiomyocytes showed that AHR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin; Dioxin (TCDD), its prototypical ligand, disrupted the expression of multiple homeobox transcription factors and inhibited cardiomyocyte contractility. Here we treated ES cells with TCDD at daily differentiation intervals to investigate whether TCDD-induced loss of contractility had a developmental window of sensitivity. Surprisingly, contractility was an AHR-dependent TCDD target solely between differentiation days 0 and 3 during the period of panmesoderm development, when TCDD also disrupted expression of genes in the TGFß/BMP2/4 and wingless-type MMTV integration site (WNT)signaling pathways, suppressed the secretion of bone morphogenetic protein (BMP4), WNT3a, and WNT5a and elevated the secretion of Activin A, as determined by ELISA of the secreted proteins in the culture medium. Supplementing the culture medium with BMP4, WNT3a, or WNT5a during the first 3 days of differentiation successfully countered TCDD-induced impairment of contractility, while anti-WNT3a, or anti-WNT5a antibodies or continuous Noggin (a BMP4 antagonist) or Activin A treatment inhibited the contractile phenotype. In Ahr(+/+), but not in Ahr(-) (/) (-) ES cells, TCDD treatment significantly increased mitochondrial copy number, suggestive of mitochondrial stress and remodeling. Sustained AHR activation during ES cell differentiation appears to disrupt the expression of signals critical to the ontogeny of cardiac mesoderm and cause the loss of contractility in the resulting cardiomyocyte lineage.


Asunto(s)
Activinas/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Células Madre Embrionarias de Ratones/citología , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Transducción de Señal/fisiología
8.
PLoS One ; 10(11): e0142440, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26555816

RESUMEN

The Developmental Origins of Health and Disease (DOHaD) Theory proposes that the environment encountered during fetal life and infancy permanently shapes tissue physiology and homeostasis such that damage resulting from maternal stress, poor nutrition or exposure to environmental agents may be at the heart of adult onset disease. Interference with endogenous developmental functions of the aryl hydrocarbon receptor (AHR), either by gene ablation or by exposure in utero to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a potent AHR ligand, causes structural, molecular and functional cardiac abnormalities and altered heart physiology in mouse embryos. To test if embryonic effects progress into an adult phenotype, we investigated whether Ahr ablation or TCDD exposure in utero resulted in cardiac abnormalities in adult mice long after removal of the agent. Ten-months old adult Ahr-/- and in utero TCDD-exposed Ahr+/+ mice showed sexually dimorphic abnormal cardiovascular phenotypes characterized by echocardiographic findings of hypertrophy, ventricular dilation and increased heart weight, resting heart rate and systolic and mean blood pressure, and decreased exercise tolerance. Underlying these effects, genes in signaling networks related to cardiac hypertrophy and mitochondrial function were differentially expressed. Cardiac dysfunction in mouse embryos resulting from AHR signaling disruption seems to progress into abnormal cardiac structure and function that predispose adults to cardiac disease, but while embryonic dysfunction is equally robust in males and females, the adult abnormalities are more prevalent in females, with the highest severity in Ahr-/- females. The findings reported here underscore the conclusion that AHR signaling in the developing heart is one potential target of environmental factors associated with cardiovascular disease.


Asunto(s)
Cardiopatías Congénitas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Animales , Femenino , Cardiopatías Congénitas/fisiopatología , Homeostasis , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Embarazo , Receptores de Hidrocarburo de Aril/genética
9.
Toxicol Sci ; 147(2): 425-35, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26139165

RESUMEN

Congenital heart disease (CHD) is the most common congenital abnormality and one of the leading causes of newborn death throughout the world. Despite much emerging scientific information, the precise etiology of this disease remains elusive. Here, we show that the aryl hydrocarbon receptor (AHR) regulates the expression of crucial cardiogenesis genes and that interference with endogenous AHR functions, either by gene ablation or by agonist exposure during early development, causes overlapping structural and functional cardiac abnormalities that lead to altered fetal heart physiology, including higher heart rates, right and left ventricle dilation, higher stroke volume, and reduced ejection fraction. With striking similarity between AHR knockout (Ahr(-/-)) and agonist-exposed wild type (Ahr(+/+)) embryos, in utero disruption of endogenous AHR functions converge into dysregulation of molecular mechanisms needed for attainment and maintenance of cardiac differentiation, including the pivotal signals regulated by the cardiogenic transcription factor NKH2.5, energy balance via oxidative phosphorylation and TCA cycle and global mitochondrial function and homeostasis. Our findings suggest that AHR signaling in the developing mammalian heart is central to the regulation of pathways crucial for cellular metabolism, cardiogenesis, and cardiac function, which are potential targets of environmental factors associated with CHD.


Asunto(s)
Corazón/embriología , Homeostasis/efectos de los fármacos , Receptores de Hidrocarburo de Aril/fisiología , Animales , Ecocardiografía , Femenino , Corazón/crecimiento & desarrollo , Homeostasis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias Cardíacas/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Transducción de Señal/fisiología
10.
J Immunol ; 194(9): 4446-57, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810390

RESUMEN

Successfully fighting infection requires a properly tuned immune system. Recent epidemiological studies link exposure to pollutants that bind the aryl hydrocarbon receptor (AHR) during development with poorer immune responses later in life. Yet, how developmental triggering of AHR durably alters immune cell function remains unknown. Using a mouse model, we show that developmental activation of AHR leads to long-lasting reduction in the response of CD8(+) T cells during influenza virus infection, cells critical for resolving primary infection. Combining genome-wide approaches, we demonstrate that developmental activation alters DNA methylation and gene expression patterns in isolated CD8(+) T cells prior to and during infection. Altered transcriptional profiles in CD8(+) T cells from developmentally exposed mice reflect changes in pathways involved in proliferation and immunoregulation, with an overall pattern that bears hallmarks of T cell exhaustion. Developmental exposure also changed DNA methylation across the genome, but differences were most pronounced following infection, where we observed inverse correlation between promoter methylation and gene expression. This points to altered regulation of DNA methylation as one mechanism by which AHR causes durable changes in T cell function. Discovering that distinct gene sets and pathways were differentially changed in developmentally exposed mice prior to and after infection further reveals that the process of CD8(+) T cell activation is rendered fundamentally different by early life AHR signaling. These findings reveal a novel role for AHR in the developing immune system: regulating DNA methylation and gene expression as T cells respond to infection later in life.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Metilación de ADN , Receptores de Hidrocarburo de Aril/genética , Virosis/genética , Virosis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Virosis/metabolismo
11.
Stem Cell Res ; 12(1): 296-308, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24316986

RESUMEN

The aryl hydrocarbon receptor (AHR) is a transcription factor and environmental sensor that regulates expression of genes involved in drug-metabolism and cell cycle regulation. Chromatin immunoprecipitation analyses, Ahr ablation in mice and studies with orthologous genes in invertebrates suggest that AHR may also play a significant role in embryonic development. To address this hypothesis, we studied the regulation of Ahr expression in mouse embryonic stem cells and their differentiated progeny. In ES cells, interactions between OCT3/4, NANOG, SOX2 and Polycomb Group proteins at the Ahr promoter repress AHR expression, which can also be repressed by ectopic expression of reprogramming factors in hepatoma cells. In ES cells, unproductive RNA polymerase II binds at the Ahr transcription start site and drives the synthesis of short abortive transcripts. Activation of Ahr expression during differentiation follows from reversal of repressive marks in Ahr promoter chromatin, release of pluripotency factors and PcG proteins, binding of Sp factors, establishment of histone marks of open chromatin, and engagement of active RNAPII to drive full-length RNA transcript elongation. Our results suggest that reversible Ahr repression in ES cells holds the gene poised for expression and allows for a quick switch to activation during embryonic development.


Asunto(s)
Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Proteínas del Grupo Polycomb/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas del Grupo Polycomb/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/metabolismo , Receptores de Hidrocarburo de Aril/genética , Factor de Transcripción Sp3 , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción
12.
Environ Health Perspect ; 121(11-12): 1334-43, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24058054

RESUMEN

BACKGROUND: The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates the expression of xenobiotic detoxification genes and is a critical mediator of gene-environment interactions. Many AHR target genes identified by genome-wide gene expression profiling have morphogenetic functions, suggesting that AHR may play a role in embryonic development. OBJECTIVES: To characterize the developmental functions of the AHR, we studied the consequences of AHR activation by the agonist 2,3,7,8-tetrachlorodibenzo-p-doxin (TCDD), and the result of its repression by the antagonists 6,2,4-trimethoxyflavone and CH 223191 or by short-hairpin RNA (shRNA)-mediated Ahr knockdown during spontaneous differentiation of embryonic stem (ES) cells into cardiomyocytes. METHODS: We generated an AHR-positive cardiomyocyte lineage differentiated from mouse ES cells that expresses puromycin resistance and enhanced green fluorescent protein (eGFP) under the control of the Cyp1a1 (cytochrome P450 1a1) promoter. We used RNA sequencing (RNA.Seq) to analyze temporal trajectories of TCDD-dependent global gene expression in these cells during differentiation. RESULTS: Activation, inhibition, and knockdown of Ahr significantly inhibited the formation of contractile cardiomyocyte nodes. Global expression analysis of AHR-positive cells showed that activation of the AHR/TCDD axis disrupted the concerted expression of genes that regulate multiple signaling pathways involved in cardiac and neural morphogenesis and differentiation, including dozens of genes encoding homeobox transcription factors and Polycomb and trithorax group proteins. CONCLUSIONS: Disruption of AHR expression levels resulted in gene expression changes that perturbed cardiomyocyte differentiation. The main function of the AHR during development appears to be the coordination of a complex regulatory network responsible for attainment and maintenance of cardiovascular homeostasis.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/fisiología , Corazón/embriología , Homeostasis/fisiología , Desarrollo de Músculos/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Compuestos Azo/farmacología , Linaje de la Célula , Resistencia a Medicamentos/fisiología , Células Madre Embrionarias/fisiología , Flavonas/farmacología , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica/fisiología , Dibenzodioxinas Policloradas/farmacología , Puromicina , Pirazoles/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo
13.
J Biol Chem ; 287(4): 2787-97, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22130668

RESUMEN

Signal transduction pathways are integral components of the developmental regulatory network that guides progressive cell fate determination. MKK4 and MKK7 are upstream kinases of the mitogen-activated protein kinases (MAPKs), responsible for channeling physiological and environmental signals to their cellular responses. Both kinases are essential for survival of mouse embryos, but because of embryonic lethality, their precise developmental roles remain largely unknown. Using gene knock-out mouse ESCs, we studied the roles of MKK4 and MKK7 in differentiation in vitro. While MKK4 and MKK7 were dispensable for ESC self-renewal and pluripotency maintenance, they exhibited unique signaling and functional properties in differentiation. MKK4 and MKK7 complemented each other in activation of the JNK-c-Jun cascades and loss of both led to senescence upon cell differentiation. On the other hand, MKK4 and MKK7 had opposite effects on activation of the p38 cascades during differentiation. Specifically, MKK7 reduced p38 activation, while Mkk7(-/-) ESCs had elevated phosphorylation of MKK4, p38, and ATF2, and increased MEF2C expression. Consequently, Mkk7(-/-) ESCs had higher expression of MHC and MLC and enhanced formation of contractile cardiomyocytes. In contrast, MKK4 was required for p38 activation and Mkk4(-/-) ESCs exhibited diminished p-ATF2 and MEF2C expression, resulting in impaired MHC induction and defective cardiomyocyte differentiation. Exogenous MKK4 expression partially restored the ability of Mkk4(-/-) ESCs to differentiate into cardiomyocytes. Our results uncover complementary and interdependent roles of MKK4 and MKK7 in development, and identify the essential requirement for MKK4 in p38 activation and cardiomyocyte differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/enzimología , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/enzimología , Células Madre Pluripotentes/enzimología , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Activación Enzimática/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 7/genética , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Fosforilación/fisiología , Células Madre Pluripotentes/citología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Gen Comp Endocrinol ; 156(2): 234-45, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18321504

RESUMEN

In the amphibian Pleurodeles waltl, estradiol treatment of genetically male larvae (ZZ) induces male-to-female sex reversal whereas heat treatment of genetically female larvae (ZW) inhibits estradiol synthesis and leads to female-to-male sex reversal. No data are available on estrogen receptors in this species. In the present study, we have isolated a unique full-length pwERalpha cDNA and its 5'-flanking region whose promoter activity was confirmed by transfection assays. RT-PCR studies performed in adult animals using ERalpha-specific primers, revealed that pwERalpha mRNA was present mainly in reproductive tissues: gonads, fat body and oviduct. PwERalpha transcript was also detected in liver, suggesting its implication in vitellogenesis control as in numerous oviparous species. The level of pwERalpha transcripts was also studied during gonad differentiation by quantitative real-time PCR. At stage 54(30d) pwERalpha expression in gonads of ZW larvae was significantly higher than in ZZ ones. This sex-specific discrimination was confirmed when gonad-mesonephros-interrenal complexes (GMI), taken at the same stage, were subjected to whole mount in situ hybridization. In comparison, the female-enriched expression of P450 aromatase, which was studied as a control of ovary differentiation, was observed earlier (stage 54). In ZW larvae reared at 32 degrees C, a condition leading to sex reversal, pwERalpha mRNA level at stage 54(30d) was lower than in control females. Taken together, these results showing a female-enriched and thermosensitive expression of pwERalpha suggest an important role for this receptor in gonad differentiation of the urodele amphibian Pleurodeles waltl.


Asunto(s)
Receptor alfa de Estrógeno/biosíntesis , Gónadas/crecimiento & desarrollo , Pleurodeles/fisiología , Maduración Sexual/fisiología , Región de Flanqueo 5'/genética , Secuencia de Aminoácidos , Animales , Línea Celular , ADN Complementario/biosíntesis , ADN Complementario/genética , Receptor alfa de Estrógeno/genética , Femenino , Calor , Hibridación in Situ , Larva/crecimiento & desarrollo , Datos de Secuencia Molecular , Ovario/crecimiento & desarrollo , Filogenia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Temperatura , Transfección
15.
Mol Reprod Dev ; 75(3): 439-49, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18008317

RESUMEN

Wild type embryos of the newt Pleurodeles waltl were used to realize parabiosis, a useful model to study the effect of endogenous circulating hormones on gonad development. The genotypic sex of each parabiont (ZZ male or ZW female) was determined early from the analysis of the sex chromosome borne marker peptidase-1. In ZZ/ZZ and ZW/ZW associations, gonads develop according to genetic sex. In ZZ/ZW associations, the ZZ gonads differentiate as normal testes while ZW gonads development shows numerous alterations. At the beginning of sex differentiation, these ZW gonads possess a reduced number of germ cells and a reduced expression of steroidogenic factor 1 and P450-aromatase mRNAs when compared to gonads from ZW/ZW associations. During gonad differentiation, conversely to the control situation, these germ cells do not enter meiosis as corroborated by chromatin status and absence of the meiosis entry marker DMC1; the activity of the estradiol-producing enzyme P450-aromatase is as low as in ZZ gonads. At adulthood, no germ cells are observed on histological sections, consistently with the absence of VASA expression. At this stage, the testis-specific marker DMRT1 is expressed only in ZZ gonads, suggesting that the somatic compartment of the ZW gonad is not masculinized. So, when exposed to ZZ hormones, ZW gonads reach the undifferentiated status but the ovary differentiation does not occur. This gonad is inhibited by a process affecting both somatic and germ cells. Additionally, the ZW gonad inhibition does not occur in the case of an exogenous estradiol treatment of larvae.


Asunto(s)
Ovario/embriología , Parabiosis , Pleurodeles/embriología , Animales , Quimerismo/embriología , Trastornos del Desarrollo Sexual/embriología , Embrión no Mamífero , Estradiol/farmacología , Femenino , Genotipo , Células Germinativas/fisiología , Masculino , Meiosis/fisiología , Modelos Biológicos , Ovario/efectos de los fármacos , Cromosomas Sexuales , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA