Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Transl Psychiatry ; 11(1): 149, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33654064

RESUMO

Maternal environmental exposures, such as high-fat diets, diabetes and obesity, can induce long-term effects in offspring. These effects include increased risk of neurodevelopmental disorders (NDDs) including autism spectrum disorder (ASD), depression and anxiety. The mechanisms underlying these late-life neurologic effects are unknown. In this article, we measured changes in the offspring brain and determined which brain regions are sensitive to maternal metabolic milieu and therefore may mediate NDD risk. We showed that mice exposed to a maternal high-fat diet display extensive brain changes in adulthood despite being switched to a low-fat diet at weaning. Brain regions impacted by early-life diet include the extended amygdalar system, which plays an important role in reward-seeking behaviour. Genes preferentially expressed in these regions have functions related to feeding behaviour, while also being implicated in human NDDs, such as autism. Our data demonstrated that exposure to maternal high-fat diet in early-life leads to brain alterations that persist into adulthood, even after dietary modifications.


Assuntos
Transtorno do Espectro Autista , Efeitos Tardios da Exposição Pré-Natal , Adulto , Filhos Adultos , Animais , Transtorno do Espectro Autista/etiologia , Encéfalo , Dieta Hiperlipídica/efeitos adversos , Feminino , Humanos , Camundongos , Gravidez
2.
J Biol Chem ; 293(22): 8449-8461, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29610276

RESUMO

The CCCTC-binding factor (CTCF) is a versatile transcriptional regulator required for embryogenesis, but its function in vascular development or in diseases with a vascular component is poorly understood. Here, we found that endothelial Ctcf is essential for mouse vascular development and limits accumulation of reactive oxygen species (ROS). Conditional knockout of Ctcf in endothelial progenitors and their descendants affected embryonic growth, and caused lethality at embryonic day 10.5 because of defective yolk sac and placental vascular development. Analysis of global gene expression revealed Frataxin (Fxn), the gene mutated in Friedreich's ataxia (FRDA), as the most strongly down-regulated gene in Ctcf-deficient placental endothelial cells. Moreover, in vitro reporter assays showed that Ctcf activates the Fxn promoter in endothelial cells. ROS are known to accumulate in the endothelium of FRDA patients. Importantly, Ctcf deficiency induced ROS-mediated DNA damage in endothelial cells in vitro, and in placental endothelium in vivo Taken together, our findings indicate that Ctcf promotes vascular development and limits oxidative stress in endothelial cells. These results reveal a function for Ctcf in vascular development, and suggest a potential mechanism for endothelial dysfunction in FRDA.


Assuntos
Fator de Ligação a CCCTC/fisiologia , Embrião de Mamíferos/patologia , Endotélio Vascular/patologia , Ataxia de Friedreich/patologia , Regulação da Expressão Gênica , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/metabolismo , Endotélio Vascular/metabolismo , Feminino , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Masculino , Camundongos , Camundongos Knockout , Frataxina
3.
Methods Mol Biol ; 1752: 11-16, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29564758

RESUMO

Whole mount immunofluorescence is a valuable technique that can be used to visualize vascular networks in early developing embryonic tissues. This technique involves the permeabilization of fixed mouse embryos and yolk sacs, and primary antibody tagging of the endothelial cell marker platelet endothelial cell adhesion molecule 1 (Pecam-1). A secondary antibody tagged with a fluorophore targets the primary antibody, fluorescently labeling endothelial cells and revealing vascular networks.


Assuntos
Embrião de Mamíferos/irrigação sanguínea , Saco Vitelino/irrigação sanguínea , Animais , Anticorpos/metabolismo , Células Endoteliais/metabolismo , Feminino , Imunofluorescência , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez
4.
PLoS Genet ; 13(8): e1006985, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28846746

RESUMO

Cardiac progenitors are specified early in development and progressively differentiate and mature into fully functional cardiomyocytes. This process is controlled by an extensively studied transcriptional program. However, the regulatory events coordinating the progression of such program from development to maturation are largely unknown. Here, we show that the genome organizer CTCF is essential for cardiogenesis and that it mediates genomic interactions to coordinate cardiomyocyte differentiation and maturation in the developing heart. Inactivation of Ctcf in cardiac progenitor cells and their derivatives in vivo during development caused severe cardiac defects and death at embryonic day 12.5. Genome wide expression analysis in Ctcf mutant hearts revealed that genes controlling mitochondrial function and protein production, required for cardiomyocyte maturation, were upregulated. However, mitochondria from mutant cardiomyocytes do not mature properly. In contrast, multiple development regulatory genes near predicted heart enhancers, including genes in the IrxA cluster, were downregulated in Ctcf mutants, suggesting that CTCF promotes cardiomyocyte differentiation by facilitating enhancer-promoter interactions. Accordingly, loss of CTCF disrupts gene expression and chromatin interactions as shown by chromatin conformation capture followed by deep sequencing. Furthermore, CRISPR-mediated deletion of an intergenic CTCF site within the IrxA cluster alters gene expression in the developing heart. Thus, CTCF mediates local regulatory interactions to coordinate transcriptional programs controlling transitions in morphology and function during heart development.


Assuntos
Cromatina/genética , Desenvolvimento Embrionário/genética , Ventrículos do Coração/crescimento & desenvolvimento , Coração/crescimento & desenvolvimento , Proteínas Repressoras/genética , Animais , Fator de Ligação a CCCTC , Diferenciação Celular/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Ventrículos do Coração/embriologia , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Organogênese/genética , Regiões Promotoras Genéticas , Ligação Proteica , Ativação Transcricional/genética
5.
Development ; 144(11): 1976-1987, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28455378

RESUMO

Defective fetoplacental vascular maturation causes intrauterine growth restriction (IUGR). A transcriptional switch initiates placental maturation, during which blood vessels elongate. However, the cellular mechanisms and regulatory pathways involved are unknown. We show that the histone methyltransferase G9a, also known as Ehmt2, activates the Notch pathway to promote placental vascular maturation. Placental vasculature from embryos with G9a-deficient endothelial progenitor cells failed to expand owing to decreased endothelial cell proliferation and increased trophoblast proliferation. Moreover, G9a deficiency altered the transcriptional switch initiating placental maturation and caused downregulation of Notch pathway effectors including Rbpj Importantly, Notch pathway activation in G9a-deficient endothelial progenitors extended embryonic life and rescued placental vascular expansion. Thus, G9a activates the Notch pathway to balance endothelial cell and trophoblast proliferation and coordinates the transcriptional switch controlling placental vascular maturation. Accordingly, G9A and RBPJ were downregulated in human placentae from IUGR-affected pregnancies, suggesting that G9a is an important regulator in placental diseases caused by defective vascular maturation.


Assuntos
Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Placenta/irrigação sanguínea , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Movimento Celular/genética , Proliferação de Células , Regulação para Baixo/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/ultraestrutura , Desenvolvimento Embrionário/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Retardo do Crescimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Organogênese/genética , Placenta/citologia , Placenta/ultraestrutura , Gravidez , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcrição Gênica , Trofoblastos/citologia , Trofoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA