Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 152(4): 873-83, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23415233

RESUMO

Embryonic stem cells (ESCs) can instruct the conversion of differentiated cells toward pluripotency following cell-to-cell fusion by a mechanism that is rapid but poorly understood. Here, we used centrifugal elutriation to enrich for mouse ESCs at sequential stages of the cell cycle and showed that ESCs in S/G2 phases have an enhanced capacity to dominantly reprogram lymphocytes and fibroblasts in heterokaryon and hybrid assays. Reprogramming success was associated with an ability to induce precocious nucleotide incorporation within the somatic partner nuclei in heterokaryons. BrdU pulse-labeling experiments revealed that virtually all successfully reprogrammed somatic nuclei, identified on the basis of Oct4 re-expression, had undergone DNA synthesis within 24 hr of fusion with ESCs. This was essential for successful reprogramming because drugs that inhibited DNA polymerase activity effectively blocked pluripotent conversion. These data indicate that nucleotide incorporation is an early and critical event in the epigenetic reprogramming of somatic cells in experimental ESC-heterokaryons.


Assuntos
Replicação do DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Linfócitos B/citologia , Fusão Celular , Núcleo Celular/metabolismo , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Humanos , Camundongos , Nucleotídeos/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo
2.
Mol Cell ; 49(6): 1023-33, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23453809

RESUMO

Genomic imprinting directs the allele-specific marking and expression of loci according to their parental origin. Differential DNA methylation at imprinted control regions (ICRs) is established in gametes and, although largely preserved through development, can be experimentally reset by fusing somatic cells with embryonic germ cell (EGC) lines. Here, we show that the Ten-Eleven Translocation proteins Tet1 and Tet2 participate in the efficient erasure of imprints in this model system. The fusion of B cells with EGCs initiates pluripotent reprogramming, in which rapid re-expression of Oct4 is accompanied by an accumulation of 5-hydroxymethylcytosine (5hmC) at several ICRs. Tet2 was required for the efficient reprogramming capacity of EGCs, whereas Tet1 was necessary to induce 5-methylcytosine oxidation specifically at ICRs. These data show that the Tet1 and Tet2 proteins have discrete roles in cell-fusion-mediated pluripotent reprogramming and imprint erasure in somatic cells.


Assuntos
Fusão Celular , Proteínas de Ligação a DNA/fisiologia , Impressão Genômica , Proteínas Proto-Oncogênicas/fisiologia , 5-Metilcitosina/análogos & derivados , Animais , Linfócitos B/citologia , Sequência de Bases , Linhagem Celular , Citosina/análogos & derivados , Citosina/metabolismo , Metilação de DNA , Dioxigenases , Células-Tronco Embrionárias/citologia , Expressão Gênica , Células Germinativas/citologia , Proteínas de Fluorescência Verde/biossíntese , Humanos , Fator de Crescimento Insulin-Like II/genética , Camundongos , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Proteínas/metabolismo , RNA Longo não Codificante/genética , Análise de Sequência de DNA
3.
Front Cell Dev Biol ; 11: 1252521, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37727506

RESUMO

Introduction: Huntington's disease (HD) remains an incurable and fatal neurodegenerative disease long after CAG-expansion mutation in the huntingtin gene (HTT) was identified as the cause. The underlying pathological mechanism, whether HTT loss of function or gain of toxicity results from mutation, remains a matter of debate. Methods: In this study, we genetically modulated wild-type or mutant HTT expression levels in isogenic human embryonic stem cells to systematically investigate their contribution to HD-specific phenotypes. Results: Using highly reproducible and quantifiable in vitro micropattern-based assays, we observed comparable phenotypes with HD mutation and HTT depletion. However, halving endogenous wild-type HTT levels did not strongly recapitulate the HD phenotypes, arguing against a classical loss of function mechanism. Remarkably, expression of CAG-expanded HTT in non-HD cells induced HD like phenotypes akin to HTT depletion. Discussion: By corollary, these results indicate a dominant negative effect of mutated HTT on its wild-type counterpart. Complementation with additional copies of wild-type HTT ameliorated the HD-associated phenotypes, strongly supporting a classical dominant negative mechanism. Understanding the molecular basis of this dominant negative effect will guide the development of efficient clinical strategies to counteract the deleterious impact of mutant HTT on the wild-type HTT function.

4.
Eur J Immunol ; 41(3): 602-10, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21246542

RESUMO

CD1 molecules present lipid antigens to T cells. An intriguing subset of human T cells recognize CD1-expressing cells without deliberately added lipids. Frequency, subset distribution, clonal composition, naïve-to-memory dynamic transition of these CD1 self-reactive T cells remain largely unknown. By screening libraries of T-cell clones, generated from CD4(+) or CD4(-) CD8(-) double negative (DN) T cells sorted from the same donors, and by limiting dilution analysis, we find that the frequency of CD1 self-reactive T cells is unexpectedly high in both T-cell subsets, in the range of 1/10-1/300 circulating T cells. These T cells predominantly recognize CD1a and CD1c and express diverse TCRs. Frequency comparisons of T-cell clones from sorted naïve and memory compartments of umbilical cord and adult blood show that CD1 self-reactive T cells are naïve at birth and undergo an age-dependent increase in the memory compartment, suggesting a naïve/memory adaptive-like population dynamics. CD1 self-reactive clones exhibit mostly Th1 and Th0 functional activities, depending on the subset and on the CD1 isotype restriction. These findings unveil the unanticipated relevance of self-lipid T-cell response in humans and clarify the basic parameters of the lipid-specific T-cell physiology.


Assuntos
Antígenos CD1/metabolismo , Subpopulações de Linfócitos T/imunologia , Imunidade Adaptativa , Adulto , Apresentação de Antígeno , Autoantígenos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Citotoxicidade Imunológica , Sangue Fetal/citologia , Sangue Fetal/imunologia , Humanos , Imunidade Celular , Memória Imunológica , Técnicas In Vitro , Recém-Nascido , Lipídeos/imunologia , Fenótipo , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/citologia
5.
Elife ; 112022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35451959

RESUMO

The Hippo pathway, a highly conserved signaling cascade that functions as an integrator of molecular signals and biophysical states, ultimately impinges upon the transcription coactivator Yes-associated protein 1 (YAP). Hippo-YAP signaling has been shown to play key roles both at the early embryonic stages of implantation and gastrulation, and later during neurogenesis. To explore YAP's potential role in neurulation, we used self-organizing neuruloids grown from human embryonic stem cells on micropatterned substrates. We identified YAP activation as a key lineage determinant, first between neuronal ectoderm and nonneuronal ectoderm, and later between epidermis and neural crest, indicating that YAP activity can enhance the effect of BMP4 stimulation and therefore affect ectodermal specification at this developmental stage. Because aberrant Hippo-YAP signaling has been implicated in the pathology of Huntington's Disease (HD), we used isogenic mutant neuruloids to explore the relationship between signaling and the disease. We found that HD neuruloids demonstrate ectopic activation of gene targets of YAP and that pharmacological reduction of YAP's transcriptional activity can partially rescue the HD phenotype.


Assuntos
Ectoderma , Doença de Huntington , Proteínas de Sinalização YAP , Proteínas de Ciclo Celular/metabolismo , Ectoderma/metabolismo , Humanos , Neurogênese , Neurulação , Transdução de Sinais/genética , Proteínas de Sinalização YAP/genética
6.
Elife ; 82019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31868585

RESUMO

Methyl-CpG-binding-Protein 2 (MeCP2) is an abundant nuclear protein highly enriched in neurons. Here we report live-cell single-molecule imaging studies of the kinetic features of mouse MeCP2 at high spatial-temporal resolution. MeCP2 displays dynamic features that are distinct from both highly mobile transcription factors and immobile histones. Stable binding of MeCP2 in living neurons requires its methyl-binding domain and is sensitive to DNA modification levels. Diffusion of unbound MeCP2 is strongly constrained by weak, transient interactions mediated primarily by its AT-hook domains, and varies with the level of chromatin compaction and cell type. These findings extend previous studies of the role of the MeCP2 MBD in high affinity DNA binding to living neurons, and identify a new role for its AT-hooks domains as critical determinants of its kinetic behavior. They suggest that limited nuclear diffusion of MeCP2 in live neurons contributes to its local impact on chromatin structure and gene expression.


Assuntos
Cromatina/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Núcleo Celular/metabolismo , Cerebelo/citologia , Cerebelo/metabolismo , DNA/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Cinética , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neurônios/citologia , Ligação Proteica , Síndrome de Rett/genética , Fatores de Transcrição/metabolismo
7.
Trends Cell Biol ; 24(2): 136-43, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24119665

RESUMO

Methylation of cytosine within DNA is associated with transcriptional repression and genome surveillance. In plants and animals, conserved pathways exist to establish and maintain this epigenetic mark. Mechanisms underlining its removal are, however, diverse and controversial and can depend on DNA synthesis (passive) or be independent of it (active). Ten-eleven translocation (Tet)-mediated conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) has recently been evoked as a possible mechanism in the initiation of active and passive DNA demethylation. This review discuses the recent progress in this exciting area.


Assuntos
Metilação de DNA , DNA/metabolismo , Animais , DNA/biossíntese , DNA/genética , Humanos
8.
Philos Trans R Soc Lond B Biol Sci ; 366(1575): 2260-5, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21727131

RESUMO

Reprogramming differentiated cells towards pluripotency can be achieved by different experimental strategies including the forced expression of specific 'inducers' and nuclear transfer. While these offer unparalleled opportunities to generate stem cells and advance disease modelling, the relatively low levels of successful reprogramming achieved (1-2%) makes a direct analysis of the molecular events associated with productive reprogramming very challenging. The generation of transient heterokaryons between human differentiated cells (such as lymphocytes or fibroblasts) and mouse pluripotent stem cell lines results in a much higher frequency of successful conversion (15% SSEA4 expressing cells) and provides an alternative approach to study early events during reprogramming. Under these conditions, differentiated nuclei undergo a series of remodelling events before initiating human pluripotent gene expression and silencing differentiation-associated genes. When combined with genetic or RNAi-based approaches and high-throughput screens, heterokaryon studies can provide important new insights into the factors and mechanisms required to reprogramme unipotent cells towards pluripotency.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Animais , Técnicas Citológicas/métodos , Humanos
9.
Cell Stem Cell ; 6(6): 547-56, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20569692

RESUMO

Embryonic stem cells (ESCs) are pluripotent, self-renewing, and have the ability to reprogram differentiated cell types to pluripotency upon cellular fusion. Polycomb-group (PcG) proteins are important for restraining the inappropriate expression of lineage-specifying factors in ESCs. To investigate whether PcG proteins are required for establishing, rather than maintaining, the pluripotent state, we compared the ability of wild-type, PRC1-, and PRC2-depleted ESCs to reprogram human lymphocytes. We show that ESCs lacking either PRC1 or PRC2 are unable to successfully reprogram B cells toward pluripotency. This defect is a direct consequence of the lack of PcG activity because it could be efficiently rescued by reconstituting PRC2 activity in PRC2-deficient ESCs. Surprisingly, the failure of PRC2-deficient ESCs to reprogram somatic cells is functionally dominant, demonstrating a critical requirement for PcG proteins in the chromatin-remodeling events required for the direct conversion of differentiated cells toward pluripotency.


Assuntos
Linfócitos B/metabolismo , Células-Tronco Embrionárias/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linfócitos B/patologia , Fusão Celular , Linhagem Celular Transformada , Reprogramação Celular/genética , Células-Tronco Embrionárias/patologia , Técnicas de Inativação de Genes , Histona-Lisina N-Metiltransferase/genética , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Células-Tronco Neoplásicas/patologia , Complexo Repressor Polycomb 2 , Proteínas do Grupo Polycomb , Proteínas Repressoras/genética , Telomerase/biossíntese , Telomerase/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
10.
Nat Cell Biol ; 12(6): 618-24, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20473294

RESUMO

Polycomb Repressor Complexes (PRCs) are important regulators of embryogenesis. In embryonic stem (ES) cells many genes that regulate subsequent stages in development are enriched at their promoters for PRC1, PRC2 and Ser 5-phosphorylated RNA Polymerase II (RNAP), and contain domains of 'bivalent' chromatin (enriched for H3K4me3; histone H3 di- or trimethylated at Lys 4 and H3K27me3; histone H3 trimethylated at Lys 27). Loss of individual PRC components in ES cells can lead to gene de-repression and to unscheduled differentiation. Here we show that Jarid2 is a novel subunit of PRC2 that is required for the co-recruitment of PRC1 and RNAP to genes that regulate development in ES cells. Jarid2-deficient ES cells showed reduced H3K4me2/me3 and H3K27me3 marking and PRC1/PRC2 recruitment, and did not efficiently establish Ser 5-phosporylated RNAP at target genes. ES cells lacking Jarid2, in contrast to previously characterized PRC1 and PRC2 mutants, did not inappropriately express PRC2 target genes. Instead, they show a severely compromised capacity for successful differentiation towards neural or mesodermal fates and failed to correctly initiate lineage-specific gene expression in vitro. Collectively, these data indicate that transcriptional priming of bivalent genes in pluripotent ES cells is Jarid2-dependent, and suggests that priming is critical for subsequent multi-lineage differentiation.


Assuntos
Cromatina/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas/metabolismo , RNA Polimerase II/metabolismo , Diferenciação Celular/genética , Histonas/genética , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes/metabolismo , Proteínas/genética , RNA Polimerase II/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA