Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 15(6): 1260-1274, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33296674

RESUMO

The multifunctional histone chaperone, SET, is essential for embryonic development in the mouse. Previously, we identified SET as a factor that is rapidly downregulated during embryonic stem cell (ESC) differentiation, suggesting a possible role in the maintenance of pluripotency. Here, we explore SET's function in early differentiation. Using immunoprecipitation coupled with protein quantitation by LC-MS/MS, we uncover factors and complexes, including P53 and ß-catenin, by which SET regulates lineage specification. Knockdown for P53 in SET-knockout (KO) ESCs partially rescues lineage marker misregulation during differentiation. Paradoxically, SET-KO ESCs show increased expression of several Wnt target genes despite reduced levels of active ß-catenin. Further analysis of RNA sequencing datasets hints at a co-regulatory relationship between SET and TCF proteins, terminal effectors of Wnt signaling. Overall, we discover a role for both P53 and ß-catenin in SET-regulated early differentiation and raise a hypothesis for SET function at the ß-catenin-TCF regulatory axis.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Chaperonas de Histonas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Chaperonas de Histonas/genética , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Proteína Supressora de Tumor p53/genética , beta Catenina/genética
2.
Stem Cell Reports ; 9(4): 1291-1303, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28966118

RESUMO

Embryonic stem cells (ESCs) are regulated by pluripotency-related transcription factors in concert with chromatin regulators. To identify additional stem cell regulators, we screened a library of endogenously labeled fluorescent fusion proteins in mouse ESCs for fluorescence loss during differentiation. We identified SET, which displayed a rapid isoform shift during early differentiation from the predominant isoform in ESCs, SETα, to the primary isoform in differentiated cells, SETß, through alternative promoters. SETα is selectively bound and regulated by pluripotency factors. SET depletion causes proliferation slowdown and perturbed neuronal differentiation in vitro and developmental arrest in vivo, and photobleaching methods demonstrate SET's role in maintaining a dynamic chromatin state in ESCs. This work identifies an important regulator of pluripotency and early differentiation, which is controlled by alternative promoter usage.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histona Acetiltransferases/genética , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proliferação de Células , Sobrevivência Celular/genética , Montagem e Desmontagem da Cromatina , Histonas/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Placa Neural/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Isoformas de Proteínas
3.
Stem Cell Reports ; 9(4): 1304-1314, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28966122

RESUMO

Embryonic stem cells (ESCs), with their dual capacity to self-renew and differentiate, are commonly used to study differentiation, epigenetic regulation, lineage choices, and more. Using non-directed retroviral integration of a YFP/Cherry exon into mouse ESCs, we generated a library of over 200 endogenously tagged fluorescent fusion proteins and present several proof-of-concept applications of this library. We show the utility of this library to track proteins in living cells; screen for pluripotency-related factors; identify heterogeneously expressing proteins; measure the dynamics of endogenously labeled proteins; track proteins recruited to sites of DNA damage; pull down tagged fluorescent fusion proteins using anti-Cherry antibodies; and test for interaction partners. Thus, this library can be used in a variety of different directions, either exploiting the fluorescent tag for imaging-based techniques or utilizing the fluorescent fusion protein for biochemical pull-down assays, including immunoprecipitation, co-immunoprecipitation, chromatin immunoprecipitation, and more.


Assuntos
Expressão Gênica , Genes Reporter , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Recombinantes de Fusão/genética , Animais , Proteínas de Transporte , Diferenciação Celular/genética , Dano ao DNA , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Heterogeneidade Genética , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Ligação Proteica
4.
EMBO Rep ; 16(12): 1609-19, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26553936

RESUMO

Pluripotent embryonic stem cells (ESCs) are characterized by distinct epigenetic features including a relative enrichment of histone modifications related to active chromatin. Among these is tri-methylation of lysine 4 on histone H3 (H3K4me3). Several thousands of the H3K4me3-enriched promoters in pluripotent cells also contain a repressive histone mark, namely H3K27me3, a situation referred to as "bivalency". While bivalent promoters are not unique to pluripotent cells, they are relatively enriched in these cell types, largely marking developmental and lineage-specific genes which are silent but poised for immediate action. The H3K4me3 and H3K27me3 modifications are catalyzed by lysine methyltransferases which are usually found within, although not entirely limited to, the Trithorax group (TrxG) and Polycomb group (PcG) protein complexes, respectively, but these do not provide selective bivalent specificity. Recent studies highlight the family of ATP-dependent chromatin remodeling proteins as regulators of bivalent domains. Here, we discuss bivalency in general, describe the machineries that catalyze bivalent chromatin domains, and portray the emerging connection between bivalency and the action of different families of chromatin remodelers, namely INO80, esBAF, and NuRD, in pluripotent cells. We posit that chromatin remodeling proteins may enable "bivalent specificity", often selectively acting on, or selectively depleted from, bivalent domains.


Assuntos
Montagem e Desmontagem da Cromatina , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Código das Histonas/genética , Animais , Epigênese Genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos , Regiões Promotoras Genéticas
5.
Genome Biol ; 16: 213, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26415775

RESUMO

BACKGROUND: Pluripotent embryonic stem cells (ESCs) have the unique ability to differentiate into every cell type and to self-renew. These characteristics correlate with a distinct nuclear architecture, epigenetic signatures enriched for active chromatin marks and hyperdynamic binding of structural chromatin proteins. Recently, several chromatin-related proteins have been shown to regulate ESC pluripotency and/or differentiation, yet the role of the major heterochromatin proteins in pluripotency is unknown. RESULTS: Here we identify Heterochromatin Protein 1ß (HP1ß) as an essential protein for proper differentiation, and, unexpectedly, for the maintenance of pluripotency in ESCs. In pluripotent and differentiated cells HP1ß is differentially localized and differentially associated with chromatin. Deletion of HP1ß, but not HP1α, in ESCs provokes a loss of the morphological and proliferative characteristics of embryonic pluripotent cells, reduces expression of pluripotency factors and causes aberrant differentiation. However, in differentiated cells, loss of HP1ß has the opposite effect, perturbing maintenance of the differentiation state and facilitating reprogramming to an induced pluripotent state. Microscopy, biochemical fractionation and chromatin immunoprecipitation reveal a diffuse nucleoplasmic distribution, weak association with chromatin and high expression levels for HP1ß in ESCs. The minor fraction of HP1ß that is chromatin-bound in ESCs is enriched within exons, unlike the situation in differentiated cells, where it binds heterochromatic satellite repeats and chromocenters. CONCLUSIONS: We demonstrate an unexpected duality in the role of HP1ß: it is essential in ESCs for maintaining pluripotency, while it is required for proper differentiation in differentiated cells. Thus, HP1ß function both depends on, and regulates, the pluripotent state.


Assuntos
Proteínas Cromossômicas não Histona/genética , Células-Tronco Embrionárias , Heterocromatina/genética , Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular/genética , Reprogramação Celular/genética , Cromatina/genética , Proteínas Cromossômicas não Histona/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Camundongos , Camundongos Knockout
6.
Cell Rep ; 10(12): 2019-31, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25818293

RESUMO

Embryonic stem cells (ESCs) possess a distinct chromatin conformation maintained by specialized chromatin proteins. To identify chromatin regulators in ESCs, we developed a simple biochemical assay named D-CAP (differential chromatin-associated proteins), using brief micrococcal nuclease digestion of chromatin, followed by liquid chromatography tandem mass spectrometry (LC-MS/MS). Using D-CAP, we identified several differentially chromatin-associated proteins between undifferentiated and differentiated ESCs, including the chromatin remodeling protein SMARCD1. SMARCD1 depletion in ESCs led to altered chromatin and enhanced endodermal differentiation. Gene expression and chromatin immunoprecipitation sequencing (ChIP-seq) analyses suggested that SMARCD1 is both an activator and a repressor and is enriched at developmental regulators and that its chromatin binding coincides with H3K27me3. SMARCD1 knockdown caused H3K27me3 redistribution and increased H3K4me3 around the transcription start site (TSS). One of the identified SMARCD1 targets was Klf4. In SMARCD1-knockdown clones, KLF4, as well as H3K4me3 at the Klf4 locus, remained high and H3K27me3 was abolished. These results propose a role for SMARCD1 in restricting pluripotency and activating lineage pathways by regulating H3K27 methylation.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Fator 4 Semelhante a Kruppel , Camundongos
7.
Methods Mol Biol ; 1042: 173-80, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23980007

RESUMO

Chromatin-protein interactions are important in determining chromosome structure and function, thereby regulating gene expression patterns. Most chromatin associated proteins bind chromatin in a transient manner, with residence times on the order of a few seconds to minutes. This is especially pertinent in mouse embryonic stem cells (ESCs), where hyperdynamic binding of chromatin associated proteins to chromatin is thought to regulate genome plasticity. In order to quantitatively measure binding dynamics of such chromatin proteins in living cells, a combination of GFP-fusion proteins and photobleaching-based assays such as fluorescence recovery after photobleaching (FRAP) and fluorescence loss in photobleaching (FLIP) are advantageous over other existing biochemical assays, because they are applied in living cells at a single cell level. In this chapter we describe a detailed protocol for performing FRAP and FLIP assays for measuring structural chromatin protein dynamics such as Heterochromatin Protein 1 (HP1) and linker histone H1 in mouse ESCs and during ESC differentiation.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Embrionárias/citologia , Recuperação de Fluorescência Após Fotodegradação/métodos , Histonas/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/análise , Clonagem Molecular , Proteínas de Fluorescência Verde/genética , Histonas/análise , Camundongos , Fotodegradação , Análise de Célula Única/métodos
8.
Nucleic Acids Res ; 41(12): 6300-15, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23630323

RESUMO

The transcriptional landscape in embryonic stem cells (ESCs) and during ESC differentiation has received considerable attention, albeit mostly confined to the polyadenylated fraction of RNA, whereas the non-polyadenylated (NPA) fraction remained largely unexplored. Notwithstanding, the NPA RNA super-family has every potential to participate in the regulation of pluripotency and stem cell fate. We conducted a comprehensive analysis of NPA RNA in ESCs using a combination of whole-genome tiling arrays and deep sequencing technologies. In addition to identifying previously characterized and new non-coding RNA members, we describe a group of novel conserved RNAs (snacRNAs: small NPA conserved), some of which are differentially expressed between ESC and neuronal progenitor cells, providing the first evidence of a novel group of potentially functional NPA RNA involved in the regulation of pluripotency and stem cell fate. We further show that minor spliceosomal small nuclear RNAs, which are NPA, are almost completely absent in ESCs and are upregulated in differentiation. Finally, we show differential processing of the minor intron of the polycomb group gene Eed. Our data suggest that NPA RNA, both known and novel, play important roles in ESCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Pequeno RNA não Traduzido/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Epigênese Genética , Histonas/genética , Masculino , Camundongos , Proteínas/genética , RNA Polimerase II/metabolismo , Splicing de RNA , Pequeno RNA não Traduzido/biossíntese , Pequeno RNA não Traduzido/fisiologia , Spliceossomos/metabolismo
9.
J Biol Chem ; 288(9): 6663-78, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23297411

RESUMO

High doses of dexamethasone (Dex) or myostatin (Mstn) induce severe atrophy of skeletal muscle. Here we show a novel microRNA1 (miR1)-mediated mechanism through which Dex promotes skeletal muscle atrophy. Using both C2C12 myotubes and mouse models of Dex-induced atrophy we show that Dex induces miR1 expression through glucocorticoid receptor (GR). We further show that Mstn treatment facilitates GR nuclear translocation and thereby induces miR1 expression. Inhibition of miR1 in C2C12 myotubes attenuated the Dex-induced increase in atrophy-related proteins confirming a role for miR1 in atrophy. Analysis of miR1 targets revealed that HSP70 is regulated by miR1 during atrophy. Our results demonstrate that increased miR1 during atrophy reduced HSP70 levels, which resulted in decreased phosphorylation of AKT, as HSP70 binds to and protects phosphorylation of AKT. We further show that loss of pAKT leads to decreased phosphorylation, and thus, enhanced activation of FOXO3, up-regulation of MuRF1 and Atrogin-1, and progression of skeletal muscle atrophy. Based on these results, we propose a model whereby Dex- and Mstn-mediated atrophic signals are integrated through miR1, which then either directly or indirectly, inhibits the proteins involved in providing protection against atrophy.


Assuntos
Anti-Inflamatórios/efeitos adversos , Dexametasona/efeitos adversos , Proteínas de Choque Térmico HSP70/metabolismo , MicroRNAs/metabolismo , Modelos Biológicos , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Células CHO , Cricetinae , Cricetulus , Dexametasona/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Choque Térmico HSP70/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Atrofia Muscular/genética , Atrofia Muscular/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Ligases SKP Culina F-Box/biossíntese , Proteínas Ligases SKP Culina F-Box/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...