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.
Stem Cells ; 31(4): 717-28, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23307555

RESUMO

The pluripotency of mouse embryonic stem cells (mESCs) is controlled by a network of transcription factors, mi-RNAs, and signaling pathways. Here, we present a new regulatory circuit that connects miR-335, Oct4, and the Retinoblastoma pathway to control mESC self-renewal and differentiation. Oct4 drives the expression of Nipp1 and Ccnf that inhibit the activity of the protein phosphatase 1 (PP1) complex to establish hyperphosphorylation of the retinoblastoma protein 1 (pRb) as a hallmark feature of self-renewing mESCs. The Oct4-Nipp1/Ccnf-PP1-pRb axis promoting mESC self-renewal is under control of miR-335 that regulates Oct4 and Rb expression. During mESC differentiation, miR-335 upregulation co-operates with the transcriptional repression of Oct4 to facilitate the collapse of the Oct4-Nipp1/Ccnf-PP1-pRb axis, pRb dephosphorylation, the exit from self-renewal, and the establishment of a pRb-regulated cell cycle program. Our results introduce Oct4-dependent control of the Rb pathway as novel regulatory circuit controlling mESC self-renewal and differentiation.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , MicroRNAs/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Western Blotting , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Células Cultivadas , Imunoprecipitação da Cromatina , Citometria de Fluxo , Imunoprecipitação , Camundongos , MicroRNAs/genética , Fator 3 de Transcrição de Octâmero/genética , Ligação Proteica , Proteína do Retinoblastoma/genética
2.
Methods Mol Biol ; 2324: 203-217, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34165717

RESUMO

There is accumulating evidence that pseudogenes can produce functionally relevant lncRNAs in a tightly controlled manner. This class of transcripts has been demonstrated to play an important role in development and disease, by controlling parental gene expression. Classically, pseudogene derived lncRNAs compete with parental transcripts for miRNAs or factors that control parental mRNA metabolisms. Recently, pseudogene lncRNAs were demonstrated to take over the control of classic chromatin modifying enzymes and alter parental gene promoter activity or genome wide gene expression. Here, we discuss a new mechanism of parental gene expression controlled by the mOct4P4 lncRNA, a sense transcript derived from the murine Oct4 pseudogene 4. mOct4P4 lncRNA specifically interacts with the RNA binding protein FUS and the Histone Methyltransferase SUV39H1 to target heterochromatin formation at the parental Oct4 promoter in trans. In addition, we will address key issues for the functional dissection of epigenetic control of parental gene promoters by pseudogene lncRNAs.


Assuntos
Cromatina/metabolismo , Epigênese Genética/genética , Regiões Promotoras Genéticas/genética , Pseudogenes/genética , RNA Longo não Codificante/genética , Alelos , Animais , Sistemas CRISPR-Cas , Proteínas do Capsídeo/metabolismo , Cromatina/genética , Cromatina/ultraestrutura , Imunoprecipitação da Cromatina , Células-Tronco Embrionárias , Regulação da Expressão Gênica , Humanos , Levivirus/genética , Metiltransferases/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo
3.
Commun Biol ; 3(1): 632, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33128015

RESUMO

The resurrection of pseudogenes during evolution produced lncRNAs with new biological function. Here we show that pseudogene-evolution created an Oct4 pseudogene lncRNA that is able to direct epigenetic silencing of the parental Oct4 gene via a 2-step, lncRNA dependent mechanism. The murine Oct4 pseudogene 4 (mOct4P4) lncRNA recruits the RNA binding protein FUS to allow the binding of the SUV39H1 HMTase to a defined mOct4P4 lncRNA sequence element. The mOct4P4-FUS-SUV39H1 silencing complex holds target site specificity for the parental Oct4 promoter and interference with individual components results in loss of Oct4 silencing. SUV39H1 and FUS do not bind parental Oct4 mRNA, confirming the acquisition of a new biological function by the mOct4P4 lncRNA. Importantly, all features of mOct4P4 function are recapitulated by the human hOCT4P3 pseudogene lncRNA, indicating evolutionary conservation. Our data highlight the biological relevance of rapidly evolving lncRNAs that infiltrate into central epigenetic regulatory circuits in vertebrate cells.


Assuntos
Metiltransferases/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Pseudogenes , RNA Longo não Codificante/genética , Proteína FUS de Ligação a RNA/genética , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular Tumoral , Epigênese Genética , Feminino , Inativação Gênica , Humanos , Metiltransferases/genética , Camundongos , Complexos Multiproteicos/genética , Neoplasias Ovarianas/genética , Regiões Promotoras Genéticas , Proteína FUS de Ligação a RNA/metabolismo , Proteínas Repressoras/genética
4.
Nat Commun ; 6: 7631, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26158551

RESUMO

Pseudogene-derived, long non-coding RNAs (lncRNAs) act as epigenetic regulators of gene expression. Here we present a panel of new mouse Oct4 pseudogenes and demonstrate that the X-linked Oct4 pseudogene Oct4P4 critically impacts mouse embryonic stem cells (mESCs) self-renewal. Sense Oct4P4 transcription produces a spliced, nuclear-restricted lncRNA that is efficiently upregulated during mESC differentiation. Oct4P4 lncRNA forms a complex with the SUV39H1 HMTase to direct the imposition of H3K9me3 and HP1α to the promoter of the ancestral Oct4 gene, located on chromosome 17, leading to gene silencing and reduced mESC self-renewal. Targeting Oct4P4 expression in primary mouse embryonic fibroblasts causes the re-acquisition of self-renewing features of mESC. We demonstrate that Oct4P4 lncRNA plays an important role in inducing and maintaining silencing of the ancestral Oct4 gene in differentiating mESCs. Our data introduces a sense pseudogene-lncRNA-based mechanism of epigenetic gene regulation that controls the cross-talk between pseudogenes and their ancestral genes.


Assuntos
Epigênese Genética/genética , Regulação da Expressão Gênica no Desenvolvimento , Metiltransferases/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Pseudogenes/genética , RNA Longo não Codificante/genética , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Autorrenovação Celular/genética , Imunoprecipitação da Cromatina , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Imunoprecipitação , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células NIH 3T3
5.
Cancer Res ; 70(17): 6925-33, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20713524

RESUMO

Loss-of-function mutations of retinoblastoma family (Rb) proteins drive tumorigenesis by overcoming barriers to cellular proliferation. Consequently, factors modulating Rb function are of great clinical import. Here, we show that miR-335 is differentially expressed in human cancer cells and that it tightly regulates the expression of Rb1 (pRb/p105) by specifically targeting a conserved sequence motif in its 3' untranslated region. We found that by altering Rb1 (pRb/p105) levels, miR-335 activates the p53 tumor suppressor pathway to limit cell proliferation and neoplastic cell transformation. DNA damage elicited an increase in miR-335 expression in a p53-dependent manner. miR-335 and p53 cooperated in a positive feedback loop to drive cell cycle arrest. Together, these results indicate that miR-335 helps control proliferation by balancing the activities of the Rb and p53 tumor suppressor pathways. Further, they establish that miR-335 activation plays an important role in the induction of p53-dependent cell cycle arrest after DNA damage.


Assuntos
MicroRNAs/genética , Neoplasias/genética , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Dano ao DNA , Retroalimentação , Humanos , Camundongos , MicroRNAs/biossíntese , Células NIH 3T3 , Neoplasias/metabolismo , Neoplasias/patologia , RNA Interferente Pequeno/genética , Proteína do Retinoblastoma/biossíntese , Proteína do Retinoblastoma/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa