Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Open ; 8(8)2019 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-31391163

RESUMO

Stem/progenitor cells are maintained by a chromatin environment, mediated in part by Polycomb group (PcG) proteins, which depress differentiation. The trophoblast-specific PcG protein SFMBT2 is known to be required for maintenance of trophoblast progenitors. Rather than binding to trophoblast-specific genes repressed in TSC, SFMBT2 is concentrated at chromocentres and regions rich in repetitive elements, specifically LINE sequences and major satellites, suggesting that it is involved in higher-order organization of the trophoblast genome. It is also found enriched at a subset of ncRNAs. Comparison of ChIP-seq datasets for other chromatin proteins reveals several stereotypical distribution patterns, suggesting that SFMBT2 interacts with several different types of chromatin complexes specific to the trophoblast lineage.

2.
Stem Cell Reports ; 10(4): 1308-1323, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29526737

RESUMO

Cooperative action of a transcription factor complex containing OCT4, SOX2, NANOG, and KLF4 maintains the naive pluripotent state; however, less is known about the mechanisms that disrupt this complex, initiating exit from pluripotency. We show that, as embryonic stem cells (ESCs) exit pluripotency, KLF4 protein is exported from the nucleus causing rapid decline in Nanog and Klf4 transcription; as a result, KLF4 is the first pluripotency transcription factor removed from transcription-associated complexes during differentiation. KLF4 nuclear export requires ERK activation, and phosphorylation of KLF4 by ERK initiates interaction of KLF4 with nuclear export factor XPO1, leading to KLF4 export. Mutation of the ERK phosphorylation site in KLF4 (S132) blocks KLF4 nuclear export, the decline in Nanog, Klf4, and Sox2 mRNA, and differentiation. These findings demonstrate that relocalization of KLF4 to the cytoplasm is a critical first step in exit from the naive pluripotent state and initiation of ESC differentiation.


Assuntos
Ciclo Celular , Núcleo Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Regulação para Baixo , Ativação Enzimática , Carioferinas/metabolismo , Fator 4 Semelhante a Kruppel , Camundongos , Células-Tronco Embrionárias Murinas , Proteína Homeobox Nanog/metabolismo , Sinais de Exportação Nuclear , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Proteína Exportina 1
3.
Cell Mol Life Sci ; 72(1): 51-72, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25194419

RESUMO

Genomic imprinting is an epigenetic gene silencing phenomenon that is specific to eutherians in the vertebrate lineage. The acquisition of both placentation and genomic imprinting has spurred interest in the possible evolutionary link for many years. In this review we examine the genetic evidence and find that while many imprinted domains are anchored by genes required for proper placenta development in a parent of origin fashion, an equal number of imprinted genes have no apparent function that depends on imprinting. Examination of recent data from studies of molecular and genetic mechanisms points to a maternal control of the selection and maintenance of imprint marks, reinforcing the importance of the oocyte in the healthy development of the placenta and fetus.


Assuntos
Evolução Biológica , Epigênese Genética , Impressão Genômica , Placentação/genética , Animais , Feminino , Humanos , Gravidez
4.
Development ; 140(22): 4480-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24154523

RESUMO

Imprinted genes play important roles in placenta development and function. Parthenogenetic embryos, deficient in paternally expressed imprinted genes, lack extra-embryonic tissues of the trophoblast lineage. Parthenogenetic trophoblast stem cells (TSCs) are extremely difficult to derive, suggesting that an imprinted gene(s) is necessary for TSC establishment or maintenance. In a candidate study, we were able to narrow the list to one known paternally expressed gene, Sfmbt2. We show that mouse embryos inheriting a paternal Sfmbt2 gene trap null allele have severely reduced placentae and die before E12.5 due to reduction of all trophoblast cell types. We infected early embryos with lentivirus vectors expressing anti-Sfmbt2 shRNAs and found that TSC derivation was significantly reduced. Together, these observations support the hypothesis that loss of SFMBT2 results in defects in maintenance of trophoblast cell types necessary for development of the extra-embryonic tissues, the placenta in particular.


Assuntos
Impressão Genômica/genética , Placentação/genética , Proteínas do Grupo Polycomb/genética , Fatores de Transcrição/genética , Trofoblastos/citologia , Alelos , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Feminino , Fertilização/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hibridização in Situ Fluorescente , Padrões de Herança/genética , Camundongos , Partenogênese/genética , Proteínas do Grupo Polycomb/metabolismo , Gravidez , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Trofoblastos/metabolismo , Inativação do Cromossomo X/genética
5.
BMC Genomics ; 12: 204, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21510876

RESUMO

BACKGROUND: The proximal region of murine Chr 2 has long been known to harbour one or more imprinted genes from classic genetic studies involving reciprocal translocations. No imprinted gene had been identified from this region until our study demonstrated that the PcG gene Sfmbt2 is expressed from the paternally inherited allele in early embryos and extraembryonic tissues. Imprinted genes generally reside in clusters near elements termed Imprinting Control Regions (ICRs), suggesting that Sfmbt2 might represent an anchor for a new imprinted domain. RESULTS: We analyzed allelic expression of approximately 20 genes within a 3.9 Mb domain and found that Sfmbt2 and an overlapping non-coding antisense transcript are the only imprinted genes in this region. These transcripts represent a very narrow imprinted gene locus. We also demonstrate that rat Sfmbt2 is imprinted in extraembryonic tissues. An interesting feature of both mouse and rat Sfmbt2 genes is the presence of a large block of miRNAs in intron 10. Other mammals, including the bovine, lack this block of miRNAs. Consistent with this association, we show that human and bovine Sfmbt2 are biallelic. Other evidence indicates that pig Sfmbt2 is also not imprinted. Further strengthening the argument for recent evolution of Sfmbt2 is our demonstration that a more distant muroid rodent, Peromyscus also lacks imprinting and the block of miRNAs. CONCLUSIONS: These observations are consistent with the hypothesis that the block of miRNAs are driving imprinting at this locus. Our results are discussed in the context of ncRNAs at other imprinted loci. Accession numbers for Peromyscus cDNA and intron 10 genomic DNA are [Genbank:HQ416417 and Genbank:HQ416418], respectively.


Assuntos
Loci Gênicos/genética , Impressão Genômica , MicroRNAs/genética , Fatores de Transcrição/genética , Alelos , Animais , Sequência de Bases , Bovinos , Metilação de DNA , Feminino , Humanos , Íntrons/genética , Camundongos , Dados de Sequência Molecular , Oócitos/metabolismo , Peromyscus/genética , Gravidez , Ratos , Proteínas Repressoras , Especificidade da Espécie
6.
Int Rev Cell Mol Biol ; 276: 215-62, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19584014

RESUMO

Genomic imprinting is an epigenetic mechanism that silences one parental allele of a small subset of genes. Many imprinted genes exhibit this property only in extraembryonic tissues-placenta and yolk sac. This has led to the idea that imprinting in mammals coevolved with some aspect of placentation. Nevertheless, many studies of imprinting have ignored the extraembryonic tissues, the yolk sac and its precursor, the primitive endoderm, in particular. The primitive endoderm is involved in very early signaling events during a critical stage in development, gastrulation, during which body plan axes and head process neuroectoderm are established. Improper signaling from primitive endoderm as a result of abnormal expression of imprinted genes has the capacity to effect long-term defects in embryonic/fetal tissues that might hitherto have been overlooked. We discuss these gaps in the knowledge, propose a mechanism for genomic imprinting based on current data, and suggest a line of investigation that will expand our understanding of this unique regulatory mechanism and its impact on development.


Assuntos
Membranas Extraembrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Animais , Mapeamento Cromossômico , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/metabolismo , Feminino , Humanos , Relações Materno-Fetais , Mutação , Filogenia , Placenta/fisiologia , Placentação/fisiologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...